EP3035961A1 - Rabies vaccine - Google Patents

Rabies vaccine

Info

Publication number
EP3035961A1
EP3035961A1 EP14755333.3A EP14755333A EP3035961A1 EP 3035961 A1 EP3035961 A1 EP 3035961A1 EP 14755333 A EP14755333 A EP 14755333A EP 3035961 A1 EP3035961 A1 EP 3035961A1
Authority
EP
European Patent Office
Prior art keywords
sequence
mrna
utr
gene
nucleic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP14755333.3A
Other languages
German (de)
French (fr)
Inventor
Margit SCHNEE
Thomas Kramps
Lothar Stitz
Benjamin Petsch
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curevac SE
Original Assignee
Curevac AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac AG filed Critical Curevac AG
Priority to EP14755333.3A priority Critical patent/EP3035961A1/en
Priority to EP18191308.8A priority patent/EP3461498A1/en
Priority to EP22151797.2A priority patent/EP4043032A1/en
Priority claimed from PCT/EP2014/002298 external-priority patent/WO2015024665A1/en
Publication of EP3035961A1 publication Critical patent/EP3035961A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/205Rhabdoviridae, e.g. rabies virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • A61K2039/552Veterinary vaccine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/20011Rhabdoviridae
    • C12N2760/20111Lyssavirus, e.g. rabies virus
    • C12N2760/20134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates to an mRNA sequence, comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof. Additionally the present invention relates to a composition comprising a plurality of mRNA sequences comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof.
  • the present invention further describes a method of treatment, postexposure prophylaxis or prophylaxis of rabies using the mRNA sequence.
  • Rabies is a viral zoonosis, endemic in more than 1 00 countries and territories (WHO epidemiological record 2007a. No. 49/50 (82):425-436.
  • Rabies vaccines WHO position paper
  • the disease is invariably fatal following the onset of clinical symptoms occurring in the absence of postexposure prophylaxis (WHO epidemiological record 2010.
  • Rabies vaccine WHO position paper). The WHO assumes 55000 rabies related deaths and the postexposure treatment (PET) of more than 1 0 million people each year (WHO Weekly epidemiological record, No. 49/50, 2007b, 82:425-436).
  • rabies vaccines include the most widely used but highly risk-prone nerve tissue vaccines, or the safer but more costly cell culture and embryonated egg vaccines (CCEEVs).
  • CEEVs cell culture and embryonated egg vaccines
  • Risks associated with nerve tissue vaccines include induction of autoimmune central nervous system disease due to their inherent myelin content; the need for multiple injections; and unreliable efficacy (Plotkin SA. Rabies. Clin Infect Dis 2000;30:4-12).
  • the WHO does not recommend the use of nerve tissue vaccines and strongly encourages the increased supply of modern and high quality vaccines to poor populations (WHO epidemiological record 2007a. No. 49/50 (82):425-436. Rabies vaccines: WHO position paper).
  • Avian embryo vaccines and cell culture vaccines contain inactivated purified virus, free from nerve protein.
  • Pre-exposure prophylaxis with cell culture vaccine is safe, and is recommended for individuals at increased risk (e.g., laboratory staff, veterinarians, animal handlers, wildlife workers and travellers to rabies-endemic areas), but it is largely restricted for reasons of cost to the developed world. Furthermore, the anti-rabies vaccine is recommended for people travelling to countries in Africa and Asia, where rabies is endemic (STI O 201 1 ).
  • the object of the underlying invention to provide an mRNA sequence coding for antigenic peptides or proteins of Rabies virus for the use as vaccine for prophylaxis or treatment of rabies, particularly for preexposure prophylaxis or postexposure prophylaxis. Furthermore it is the object of the present invention to provide an effective rabies vaccine which can be stored without cold chain and which enables rapid and scalable vaccine production.
  • the objects underlying the present invention are solved according to a first aspect by an inventive mRNA sequence comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof.
  • the immune system may protect organisms from infection. If a pathogen breaks through a physical barrier of an organism and enters this organism, the innate immune system provides an immediate, but non-specific response. If pathogens evade this innate response, vertebrates possess a second layer of protection, the adaptive immune system. Here, the immune system adapts its response during an infection to improve its recognition of the pathogen. This improved response is then retained after the pathogen has been eliminated, in the form of an immunological memory, and allows the adaptive immune system to mount faster and stronger attacks each time this pathogen is encountered. According to this, the immune system comprises the innate and the adaptive immune system. Each of these two parts contains so called humoral and cellular components.
  • Immune response An immune response may typically either be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response).
  • the invention relates to the core to specific reactions (adaptive immune responses) of the adaptive immune system. Particularly, it relates to adaptive immune responses to infections by viruses like e.g. Rabies. However, this specific response can be supported by an additional unspecific reaction (innate immune response). Therefore, the invention also relates to a compound for simultaneous stimulation of the innate and the adaptive immune system to evoke an efficient adaptive immune response.
  • Adaptive immune system The adaptive immune system is composed of highly specialized, systemic cells and processes that eliminate or prevent pathogenic growth.
  • the adaptive immune response provides the vertebrate immune system with the ability to recognize and remember specific pathogens (to generate immunity), and to mount stronger attacks each time the pathogen is encountered.
  • the system is highly adaptable because of somatic hypermutation (a process of increased frequency of somatic mutations), and V(D)J recombination (an irreversible genetic recombination of antigen receptor gene segments). This mechanism allows a small number of genes to generate a vast number of different antigen receptors, which are then uniquely expressed on each individual lymphocyte.
  • Immune network theory is a theory of how the adaptive immune system works, that is based on interactions between the variable regions of the receptors of T cells, B cells and of molecules made by T cells and B cells that have variable regions.
  • Adaptive immune response is typically understood to be antigen-specific. Antigen specificity allows for the generation of responses that are tailored to specific antigens, pathogens or pathogen-infected cells. The ability to mount these tailored responses is maintained in the body by "memory cells". Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it.
  • the first step of an adaptive immune response is the activation of naive antigen- specific T cells or different immune cells able to induce an antigen-specific immune response by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • Dendritic cells that can serve as antigen-presenting cells are inter alia dendritic cells, macrophages, and B cells. Each of these cells has a distinct function in eliciting immune responses.
  • Dendritic cells take up antigens by phagocytosis and macropinocytosis and are stimulated by contact with e.g. a foreign antigen to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells.
  • Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents or other appropriate stimuli to express MHC molecules.
  • the unique ability of B cells to bind and internalize soluble protein antigens via their receptors may also be important to induce T cells.
  • T cells which induces their proliferation and differentiation into armed effector T cells.
  • the most important function of effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by Th1 cells which together make up cell-mediated immunity, and the activation of B cells by both Th2 and Th1 cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cell receptors which do not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogen-derived protein antigens, which are bound to MHC molecules on the surfaces of other cells.
  • Cellular immunity/cellular immune response relates typically to the activation of macrophages, natural killer cells (NK), antigen-specific cytotoxic T- lymphocytes, and the release of various cytokines in response to an antigen.
  • cellular immunity is not related to antibodies but to the activation of cells of the immune system.
  • a cellular immune response is characterized e.g.
  • Humoral immunity/humoral immune response Humoral immunity refers typically to antibody production and the accessory processes that may accompany it.
  • a humoral immune response may be typically characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation.
  • Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
  • the innate immune system also known as non-specific immune system, comprises the cells and mechanisms that defend the host from infection by other organisms in a non-specific manner. This means that the cells of the innate system recognize and respond to pathogens in a generic way, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host.
  • the innate immune system may be e.g. activated by ligands of pathogen-associated molecular patterns (PAMP) receptors, e.g.
  • PAMP pathogen-associated molecular patterns
  • TLRs Toll-like receptors
  • auxiliary substances such as lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-1 0, IL-12, IL- 13, IL-14, IL-1 5, IL-16, IL-1 7, IL-1 8, IL-1 9, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL- 27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G- CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and
  • a response of the innate immune system includes recruiting immune cells to sites of infection, through the production of chemical factors, including specialized chemical mediators, called cytokines; activation of the complement cascade; identification and removal of foreign substances present in organs, tissues, the blood and lymph, by specialized white blood cells; activation of the adaptive immune system through a process known as antigen presentation; and/or acting as a physical and chemical barrier to infectious agents.
  • Adjuvant / adjuvant component An adjuvant or an adjuvant component in the broadest sense is typically a (e.g. pharmacological or immunological) agent or composition that may modify, e.g. enhance, the efficacy of other agents, such as a drug or vaccine.
  • a (e.g. pharmacological or immunological) agent or composition that may modify, e.g. enhance, the efficacy of other agents, such as a drug or vaccine.
  • the term refers in the context of the invention to a compound or composition that serves as a carrier or auxiliary substance for immunogens and/or other pharmaceutically active compounds. It is to be interpreted in a broad sense and refers to a broad spectrum of substances that are able to increase the immunogenicity of antigens incorporated into or coadministered with an adjuvant in question.
  • an adjuvant will preferably enhance the specific immunogenic effect of the active agents of the present invention.
  • adjuvant or “adjuvant component” has the same meaning and can be used mutually.
  • Adjuvants may be divided, e.g., into immuno potentiators, antigenic delivery systems or even combinations thereof.
  • adjuvant is typically understood not to comprise agents which confer immunity by themselves.
  • An adjuvant assists the immune system unspecifically to enhance the antigen-specific immune response by e.g. promoting presentation of an antigen to the immune system or induction of an unspecific innate immune response.
  • an adjuvant may preferably e.g. modulate the antigen-specific immune response by e.g. shifting the dominating Th2-based antigen specific response to a more Th1 -based antigen specific response or vice versa. Accordingly, an adjuvant may favourably modulate cytokine expression/secretion, antigen presentation, type of immune response etc.
  • Immunostimulatory RNA in the context of the invention may typical ly be a RNA that is able to induce an innate immune response itself. It usually does not have an open reading frame and thus does not provide a peptide-antigen or immunogen but elicits an innate immune response e.g. by binding to a specific kind of Tolllike-receptor (TLR) or other suitable receptors. However, of course also mRNAs having an open reading frame and coding for a peptide/protein (e.g. an antigenic function) may induce an i nnate immune response.
  • TLR Tolllike-receptor
  • the term "antigen" refers typically to a substance which may be recognized by the immune system and may be capable of triggering an antigen-specific immune response, e.g. by formation of antibodies or antigen- specific T-cells as part of an adaptive immune response.
  • An antigen may be a protein or peptide.
  • the first step of an adaptive immune response is the activation of naive antigen-specific T cells by antigen-presenting cel ls. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • the three cel l types that can serve as antigen-presenting cel ls are dendritic cells, macrophages, and B cel ls.
  • Tissue dendritic cel ls take up antigens by phagocytosis and macropinocytosis and are stimulated by infection to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells.
  • Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents to express MHC class II molecules.
  • the unique ability of B cells to bind and internalize soluble protein antigens via their receptors may be important to induce T cells. By presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells.
  • effector T cells The most important function of effector T cells is the killing of infected cells by CD8 + cytotoxic T cells and the activation of macrophages by TH1 cells which together make up cell-mediated immunity, and the activation of B cells by both TH2 and TH 1 cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cel l receptors which does not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogens' protein antigens, which are bound to MHC molecules on the surfaces of other cel ls.
  • T cells fall into two major classes that have different effector functions. The two classes are distinguished by the expression of the cell-surface proteins CD4 and CD8.
  • T cells differ in the class of MHC molecule that they recognize.
  • MHC molecules There are two classes of MHC molecules - MHC class I and MHC class II molecules - which differ in their structure and expression pattern on tissues of the body.
  • CD4 + T cells bind to a MHC class II molecule and CD8 + T cells to a MHC class I molecule.
  • MHC class I and MHC class II molecules have distinct distributions among cells that reflect the different effector functions of the T cells that recognize them.
  • MHC class I molecules present peptides of cytosolic and nuclear origin e.g. from pathogens, commonly viruses, to CD8 + T cells, which differentiate into cytotoxic T cells that are specialized to kill any cell that they specifically recognize.
  • MHC class I molecules Almost all cells express MHC class I molecules, although the level of constitutive expression varies from one cell type to the next. But not only pathogenic peptides from viruses are presented by MHC class I molecules, also self-antigens like tumour antigens are presented by them. MHC class I molecules bind peptides from proteins degraded in the cytosol and transported in the endoplasmic reticulum. The CD8 + T cells that recognize MHC class l:peptide complexes at the surface of infected cells are specialized to kill any cells displaying foreign peptides and so rid the body of cells infected with viruses and other cytosolic pathogens.
  • CD4 + T cells CD4 + helper T cells
  • MHC class II molecules are normally found on B lymphocytes, dendritic cells, and macrophages, cells that participate in immune responses, but not on other tissue cells. Macrophages, for example, are activated to kill the intravesicular pathogens they harbour, and B cells to secrete immunoglobulins against foreign molecules. MHC class II molecules are prevented from binding to peptides in the endoplasmic reticulum and thus MHC class II molecules bind peptides from proteins which are degraded in endosomes.
  • TH1 cells can capture peptides from pathogens that have entered the vesicular system of macrophages, or from antigens internalized by immature dendritic cells or the immunoglobulin receptors of B cells.
  • Pathogens that accumulate in large numbers inside macrophage and dendritic cell vesicles tend to stimulate the differentiation of TH1 cells, whereas extracellular antigens tend to stimulate the production of TH2 cells.
  • TH1 cells activate the microbicidal properties of macrophages and induce B cells to make lgG antibodies that are very effective of opsonising extracellular pathogens for ingestion by phagocytic cells
  • TH2 cells initiate the humoral response by activating naive B cells to secrete IgM, and induce the production of weakly opsonising antibodies such as lgG1 and lgG3 (mouse) and lgG2 and lgG4 (human) as well as IgA and IgE (mouse and human).
  • T cell epitopes or parts of the proteins in the context of the present invention may comprise fragments preferably having a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 1 3 or more amino acids, e.g. 1 3, 14, 15, 1 6, 1 7, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T cells in form of a complex consisting of the peptide fragment and an MHC molecule.
  • B cell epitopes are typically fragments located on the outer surface of (native) protein or peptide antigens as defined herein, preferably having 5 to 1 5 amino acids, more preferably having 5 to 12 amino acids, even more preferably having 6 to 9 amino acids, which may be recognized by antibodies, i.e. in their native form.
  • Such epitopes of proteins or peptides may furthermore be selected from any of the herein mentioned variants of such proteins or peptides.
  • antigenic determinants can be conformational or discontinuous epitopes which are composed of segments of the proteins or peptides as defined herein that are discontinuous in the amino acid sequence of the proteins or peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain.
  • Vaccine A vaccine is typically understood to be a prophylactic or therapeutic material providing at least one antigen or antigenic function. The antigen or antigenic function may stimulate the body's adaptive immune system to provide an adaptive immune response.
  • Antigen-providing mRNA in the context of the invention may typically be an mRNA, having at least one open reading frame that can be translated by a cell or an organism provided with that mRNA.
  • the product of this translation is a peptide or protein that may act as an antigen, preferably as an immunogen.
  • the product may also be a fusion protein composed of more than one immunogen, e.g. a fusion protein that consist of two or more epitopes, peptides or proteins derived from the same or different virus-proteins, wherein the epitopes, peptides or proteins may be linked by linker sequences.
  • Bi-/multicistronic mRNA that typically may have two (bicistronic) or more (multicistronic) open reading frames (ORF).
  • An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein. Translation of such a mRNA yields two (bicistronic) or more (multicistronic) distinct translation products (provided the ORFs are not identical).
  • such mRNAs may for example comprise an internal ribosomal entry site (IRES) sequence.
  • a 5'-CAP is typically a modified nucleotide, particularly a guanine nucleotide, added to the 5' end of an mRNA-molecule.
  • the 5'-CAP is added using a 5 '-5 '-triphosphate linkage (also named m7GpppN).
  • 5'-CAP structures include glyceryl, inverted deoxy abasic residue (moiety), 4', 5' methylene nucleotide, l -(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, 1 ,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4- dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3 '-3 '-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3 '-2 '-inverted nucleotide moiety, 3
  • modified 5'-CAP structures may be used in the context of the present invention to modify the inventive mRNA sequence.
  • Further modified 5'-CAP structures which may be used in the context of the present invention are CAP1 (methylation of the ribose of the adjacent nucleotide of m7GpppN), CAP2 (methylation of the ribose of the 2 nd nucleotide downstream of the m7GpppN), CAP3 (methylation of the ribose of the 3 rd nucleotide downstream of the m7GpppN), CAP4 (methylation of the ribose of the 4 th nucleotide downstream of the m7GpppN), ARCA (anti- reverse CAP analogue, modified ARCA (e.g.
  • Fragments of proteins in the context of the present invention may, typically, comprise a sequence of a protein or peptide as defined herein, which is, with regard to its amino acid sequence (or its encoded nucleic acid molecule), N- terminally and/or C-terminally truncated compared to the amino acid sequence of the original (native) protein (or its encoded nucleic acid molecule). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level.
  • a sequence identity with respect to such a fragment as defined herein may therefore preferably refer to the entire protein or peptide as defined herein or to the entire (coding) nucleic acid molecule of such a protein or peptide.
  • Fragments of proteins or peptides in the context of the present invention may furthermore comprise a sequence of a protein or peptide as defined herein, which has a length of for example at least 5 amino acids, preferably a length of at least 6 amino acids, preferably at least 7 amino acids, more preferably at least 8 amino acids, even more preferably at least 9 amino acids; even more preferably at least 10 amino acids; even more preferably at least 1 1 amino acids; even more preferably at least 12 amino acids; even more preferably at least 13 amino acids; even more preferably at least 14 amino acids; even more preferably at least 1 5 amino acids; even more preferably at least 1 6 amino acids; even more preferably at least 1 7 amino acids; even more preferably at least 1 8 amino acids; even more preferably at least 19 amino acids; even more preferably at least 20 amino acids; even more preferably at least 25 amino acids; even more preferably at least 30 amino acids; even more preferably at least 35 amino acids; even more preferably at least 50 amino acids; or most preferably at least 100 amino acids
  • such fragment may have a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 1 3 or more amino acids, e.g. 13, 14, 1 5, 1 6, 1 7, 1 8, 1 9, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e.
  • fragments are typically not recognized in their native form. Fragments of proteins or peptides may comprise at least one epitope of those proteins or peptides. Furthermore also domains of a protein, like the extracellular domain, the intracellular domain or the transmembrane domain and shortened or truncated versions of a protein may be understood to comprise a fragment of a protein.
  • Variants of proteins may be generated, having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s). Preferably, these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property. "Variants” of proteins or peptides as defined in the context of the present invention may comprise conservative amino acid substitution(s) compared to their native, i.e. non-mutated physiological, sequence. Those amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined herein.
  • amino acids which originate from the same class, are exchanged for one another are called conservative substitutions.
  • these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function.
  • an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g.
  • Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1 985, Absorption, Circular Dichroism and ORD of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger et a/, (ed.), Elsevier, Amsterdam).
  • a "variant" of a protein or peptide may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid identity over a stretch of 10, 20, 30, 50, 75 or 100 amino acids of such protein or peptide.
  • variants of proteins or peptides as defined herein, which may be encoded by a nucleic acid molecule may also comprise those sequences, wherein nucleotides of the encoding nucleic acid sequence are exchanged according to the degeneration of the genetic code, without leading to an alteration of the respective amino acid sequence of the protein or peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutation(s) within the above meaning.
  • Identity of a sequence In order to determine the percentage to which two sequences are identical, e.g.
  • nucleic acid sequences or amino acid sequences as defined herein preferably the amino acid sequences encoded by a nucleic acid sequence of the polymeric carrier as defined herein or the amino acid sequences themselves, the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. a position of a first sequence may be compared with the corresponding position of the second sequence. If a position in the first sequence is occupied by the same component (residue) as is the case at a position in the second sequence, the two sequences are identical at this position. If this is not the case, the sequences differ at this position. If insertions occur in the second sequence in comparison to the first sequence, gaps can be inserted into the first sequence to allow a further alignment.
  • a position of a first sequence may be compared with the corresponding position of the second sequence. If a position in the first sequence is occupied by the same component (residue) as is the case at a position in the second sequence, the two sequences are
  • deletions occur in the second sequence in comparison to the first sequence, gaps can be inserted into the second sequence to allow a further alignment.
  • the percentage to which two sequences are identical is then a function of the number of identical positions divided by the total number of positions including those positions which are only occupied in one sequence.
  • the percentage to which two sequences are identical can be determined using a mathematical algorithm.
  • a preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. (1 993), PNAS USA, 90:5873-5877 or Altschul et al. (1 997), Nucleic Acids Res., 25:3389-3402. Such an algorithm is integrated in the BLAST program. Sequences which are identical to the sequences of the present invention to a certain extent can be identified by this program.
  • a derivative of a peptide or protein is typically understood to be a molecule that is derived from another molecule, such as said peptide or protein.
  • a "derivative" of a peptide or protein also encompasses fusions comprising a peptide or protein used in the present invention.
  • the fusion comprises a label, such as, for example, an epitope, e.g., a FLAG epitope or a V5 epitope.
  • the epitope is a FLAG epitope.
  • a tag is useful for, for example, purifying the fusion protein.
  • a monocistronic mRNA may typically be an mRNA, that encodes only one open reading frame.
  • An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein.
  • nucleic acid means any DNA- or RNA-molecule and is used synonymous with polynucleotide. Wherever herein reference is made to a nucleic acid or nucleic acid sequence encoding a particular protein and/or peptide, said nucleic acid or nucleic acid sequence, respectively, preferably also comprises regulatory sequences allowing in a suitable host, e.g. a human being, its expression, i.e. transcription and/or translation of the nucleic acid sequence encoding the particular protein or peptide.
  • a suitable host e.g. a human being
  • a peptide is a polymer of amino acid monomers. Usually the monomers are linked by peptide bonds.
  • the term "peptide" does not limit the length of the polymer chain of amino acids. In some embodiments of the present invention a peptide may for example contain less than 50 monomer units. Longer peptides are also called polypeptides, typically having 50 to 600 monomeric units, more specifically 50 to 300 monomeric units.
  • Pharmaceutically effective amount A pharmaceutically effective amount in the context of the invention is typically understood to be an amount that is sufficient to induce an immune response.
  • a protein typically consists of one or more peptides and/or polypeptides folded into 3-dimensional form, facilitating a biological function.
  • a poly-(C)-sequence is typically a long sequence of cytosine nucleotides, typically about 10 to about 200 cytosine nucleotides, preferably about 10 to about 100 cytosine nucleotides, more preferably about 10 to about 70 cytosine nucleotides or even more preferably about 20 to about 50 or even about 20 to about 30 cytosine nucleotides.
  • a poly(C) sequence may preferably be located 3' of the coding region comprised by a nucleic acid.
  • Poly-A-tail A poly-A-tail also called "3'-poly(A) tail” is typically a long sequence of adenosine nucleotides of up to about 400 adenosine nucleotides, e.g. from about 25 to about 400, preferably from about 50 to about 400, more preferably from about 50 to about 300, even more preferably from about 50 to about 250, most preferably from about 60 to about 250 adenosine nucleotides, added to the 3' end of a RNA.
  • Stabilized nucleic acid A stabilized nucleic acid, typically, exhibits a modification increasing resistance to in vivo degradation (e.g.
  • RNA can, e.g., be achieved by providing a 5'-CAP- Structure, a Poly-A-Tail, or any other UTR-modification. It can also be achieved by backbone- modification or modification of the G/C-content of the nucleic acid.
  • Stabilization of RNA can, e.g., be achieved by providing a 5'-CAP- Structure, a Poly-A-Tail, or any other UTR-modification. It can also be achieved by backbone- modification or modification of the G/C-content of the nucleic acid.
  • Various other methods are known in the art and conceivable in the context of the invention.
  • Carrier / polymeric carrier A carrier in the context of the invention may typically be a compound that facilitates transport and/or complexation of another compound. Said carrier may form a complex with said other compound.
  • a polymeric carrier is a carrier that is formed of a polymer.
  • Cationic component typically refers to a charged molecule, which is positively charged (cation) at a pH value of typically about 1 to 9, preferably of a pH value of or below 9 (e.g. 5 to 9), of or below 8 (e.g. 5 to 8), of or below 7 (e.g. 5 to 7), most preferably at physiological pH values, e.g. about 7.3 to 7.4. Accordingly, a cationic peptide, protein or polymer according to the present invention is positively charged under physiological conditions, particularly under physiological salt conditions of the cell in vivo.
  • a cationic peptide or protein preferably contains a larger number of cationic amino acids, e.g.
  • cationic may also refer to "polycationic" components.
  • 3'-untranslated region A 3'UTR is typically the part of an mRNA which is located between the protein coding region (i.e. the open reading frame) and the poly(A) sequence of the mRNA. A 3'UTR of the mRNA is not translated into an amino acid sequence.
  • the 3'UTR sequence is generally encoded by the gene which is transcribed into the respective mRNA during the gene expression process. The genomic sequence is first transcribed into pre-mature mRNA, which comprises optional introns.
  • the pre-mature mRNA is then further processed into mature mRNA in a maturation process.
  • This maturation process comprises the steps of 5'-Capping, splicing the pre-mature mRNA to excise optional introns and modifications of the 3'-end, such as polyadenylation of the 3'-end of the premature mRNA and optional endo- or exonuclease cleavages etc.
  • a 3'UTR corresponds to the sequence of a mature mRNA which is located 3' to the stop codon of the protein coding region, preferably immediately 3' to the stop codon of the protein coding region, and which extends to the 5'-side of the poly(A) sequence, preferably to the nucleotide immediately 5' to the poly(A) sequence.
  • the term "corresponds to" means that the 3'UTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining the 3'UTR sequence, or a DNA sequence which corresponds to such RNA sequence.
  • a 3'UTR of a gene such as “a 3'UTR of an albumin gene” is the sequence which corresponds to the 3'UTR of the mature mRNA derived from this gene, i.e. the mRNA obtained by transcription of the gene and maturation of the pre-mature mRNA.
  • the term "3'UTR of a gene” encompasses the DNA sequence and the RNA sequence of the 3'UTR.
  • a 5'-untranslated region is typically understood to be a particular section of messenger RNA (mRNA). It is located 5' of the open reading frame of the mRNA. Typically, the 5'UTR starts with the transcriptional start site and ends one nucleotide before the start codon of the open reading frame.
  • the 5'-UTR may comprise elements for controlling gene expression, also called regulatory elements. Such regulatory elements may be, for example, ribosomal binding sites or a 5'-Terminal Oligopyrimidine Tract.
  • the 5'UTR may be posttranscriptionally modified, for example by addition of a 5'-CAP.
  • a 5'UTR corresponds to the sequence of a mature mRNA which is located between the 5'-CAP and the start codon.
  • the 5'UTR corresponds to the sequence which extends from a nucleotide located 3' to the 5'-CAP, preferably from the nucleotide located immediately 3' to the 5'-CAP, to a nucleotide located 5' to the start codon of the protein coding region, preferably to the nucleotide located immediately 5' to the start codon of the protein coding region.
  • the nucleotide located immediately 3' to the 5'-CAP of a mature mRNA typically corresponds to the transcriptional start site.
  • the term “corresponds to” means that the 5'UTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining the 5'UTR sequence, or a DNA sequence which corresponds to such RNA sequence.
  • a 5'UTR of a gene such as "a 5'UTR of a TOP gene” is the sequence which corresponds to the 5'UTR of the mature mRNA derived from this gene, i.e. the mRNA obtained by transcription of the gene and maturation of the pre-mature mRNA.
  • the term “5'UTR of a gene” encompasses the DNA sequence and the RNA sequence of the 5'UTR.
  • TOP 5'Terminal Oligopyrimidine Tract
  • the 5'terminal oligopyrimidine tract (TOP) is typically a stretch of pyrimidine nucleotides located at the 5' terminal region of a nucleic acid molecule, such as the 5' terminal region of certain mRNA molecules or the 5' terminal region of a functional entity, e.g. the transcribed region, of certain genes.
  • the sequence starts with a cytidine, which usually corresponds to the transcriptional start site, and is followed by a stretch of usually about 3 to 30 pyrimidine nucleotides.
  • the TOP may comprise 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 1 3, 14, 1 5, 1 6, 1 7, 18, 1 9, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30 or even more nucleotides.
  • Messenger RNA that contains a 5'terminal oligopyrimidine tract is often referred to as TOP mRNA. Accordingly, genes that provide such messenger RNAs are referred to as TOP genes.
  • TOP sequences have, for example, been found in genes and mRNAs encoding peptide elongation factors and ribosomal proteins.
  • TOP motif In the context of the present invention, a TOP motif is a nucleic acid sequence which corresponds to a 5'TOP as defined above. Thus, a TOP motif in the context of the present invention is preferably a stretch of pyrimidine nucleotides having a length of 3-30 nucleotides.
  • the TOP-motif consists of at least 3 pyrimidine nucleotides, preferably at least 4 pyrimidine nucleotides, preferably at least 5 pyrimidine nucleotides, more preferably at least 6 nucleotides, more preferably at least 7 nucleotides, most preferably at least 8 pyrimidine nucleotides, wherein the stretch of pyrimidine nucleotides preferably starts at its 5'end with a cytosine nucleotide.
  • the TOP-motif preferably starts at its 5'end with the transcriptional start site and ends one nucleotide 5' to the first purin residue in said gene or mRNA.
  • a TOP motif in the sense of the present invention is preferably located at the 5'end of a sequence which represents a 5'UTR or at the 5'end of a sequence which codes for a 5'UTR.
  • TOP motif a stretch of 3 or more pyrimidine nucleotides is called "TOP motif" in the sense of the present invention if this stretch is located at the 5'end of a respective sequence, such as the inventive mRNA, the 5'UTR element of the inventive mRNA, or the nucleic acid sequence which is derived from the 5'UTR of a TOP gene as described herein.
  • a stretch of 3 or more pyrimidine nucleotides which is not located at the 5'-end of a 5'UTR or a 5'UTR element but anywhere within a 5'UTR or a 5'UTR element is preferably not referred to as "TOP motif".
  • TOP genes are typically characterised by the presence of a 5' terminal oligopyrimidine tract. Furthermore, most TOP genes are characterized by a growth- associated translational regulation. However, also TOP genes with a tissue specific translational regulation are known. As defined above, the 5'UTR of a TOP gene corresponds to the sequence of a 5'UTR of a mature mRNA derived from a TOP gene, which preferably extends from the nucleotide located 3' to the 5'-CAP to the nucleotide located 5' to the start codon. A 5'UTR of a TOP gene typically does not comprise any start codons, preferably no upstream AUGs (uAUGs) or upstream open reading frames (uORFs).
  • uAUGs upstream AUGs
  • uORFs upstream open reading frames
  • upstream AUGs and upstream open reading frames are typically understood to be AUGs and open reading frames that occur 5' of the start codon (AUG) of the open reading frame that should be translated.
  • the 5'UTRs of TOP genes are generally rather short.
  • the lengths of 5'UTRs of TOP genes may vary between 20 nucleotides up to 500 nucleotides, and are typically less than about 200 nucleotides, preferably less than about 1 50 nucleotides, more preferably less than about 100 nucleotides.
  • Exemplary 5'UTRs of TOP genes in the sense of the present invention are the nucleic acid sequences extending from the nucleotide at position 5 to the nucleotide located immediately 5' to the start codon (e.g.
  • a particularly preferred fragment of a 5'UTR of a TOP gene is a 5'UTR of a TOP gene lacking the 5'TOP motif.
  • the term '5'UTR of a TOP gene' preferably refers to the 5'UTR of a naturally occurring TOP gene.
  • a fragment of a nucleic acid sequence consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length nucleic acid sequence which is the basis for the nucleic acid sequence of the fragment, which represents at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length nucleic acid sequence.
  • Such a fragment in the sense of the present invention, is preferably a functional fragment of the full-length nucleic acid sequence.
  • a variant of a nucleic acid sequence refers to a variant of nucleic acid sequences which forms the basis of a nucleic acid sequence.
  • a variant nucleic acid sequence may exhibit one or more nucleotide deletions, insertions, additions and/or substitutions compared to the nucleic acid sequence from which the variant is derived.
  • a variant of a nucleic acid sequence is at least 40%, preferably at least 50%, more preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, even more preferably at least 90%, most preferably at least 95% identical to the nucleic acid sequence the variant is derived from.
  • the variant is a functional variant.
  • a "variant" of a nucleic acid sequence may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% nucleotide identity over a stretch of 1 0, 20, 30, 50, 75 or 1 00 nucleotide of such nucleic acid sequence.
  • homolog of a nucleic acid sequence refers to sequences of other species than the particular sequence. It is particular preferred that the nucleic acid sequence is of human origin and therefore it is preferred that the homolog is a homolog of a human nucleic acid sequence.
  • Jet injection refers to a needle-free injection method, wherein a fluid containing at least one inventive mRNA sequence and, optional ly, further suitable excipients is forced through an orifice, thus generating an ultra-fine liquid stream of high pressure that is capable of penetrating mammalian skin and, depending on the injection settings, subcutaneous tissue or muscle tissue.
  • the liquid stream forms a hole in the skin, through which the liquid stream is pushed into the target tissue.
  • jet injection is used for intradermal, subcutaneous or intramuscular injection of the mRNA sequence according to the invention.
  • jet injection is used for intramuscular injection of the mRNA sequence according to the invention.
  • jet injection is used for intradermal injection of the mRNA sequence according to the invention.
  • the present invention is based on the surprising finding of the present inventors that an mRNA sequence comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus induces antigen-specific immune responses which neutralize Rabies virus particles and therefore prevent Rabies virus infections. It was very surprising for the inventors that the inventive mRNA sequence induces at least the same immune responses than a licensed rabies vaccine which consists of the whole inactived rabies virus. Even more surprisingly the inventive mRNA sequence coding for an antigenic protein of Rabies virus induced higher antigen-specific CD4+-T cells than a licenced rabies vaccine.
  • the inventive mRNA sequence comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus could contribute to affordable, readily available, temperature-stable rabies vaccines, " particularly for preexposure and postexposure rabies prophylaxis for the developed and developing world.
  • the mRNA sequence according to the invention enables rapid and rational vaccine design with flexibility, speed and scalability of production probably exceeding those of current virus-based technologies.
  • the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof.
  • the coding region of the inventive mRNA sequence according to the first aspect of the present invention may occur as a mono-, di-, or even multicistronic mRNA, i.e. an mRNA sequence which carries the coding sequences of one, two or more proteins or peptides.
  • Such coding sequences in di-, or even multicistronic mRNAs may be separated by at least one internal ribosome entry site (IRES) sequence, e.g. as described herein or by signal peptides which induce the cleavage of the resulting polypeptide which comprises several proteins or peptides.
  • IRS internal ribosome entry site
  • the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof.
  • the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G) of Rabies virus or a fragment, variant or derivative thereof.
  • RAV-G glycoprotein G
  • amino acid sequence of the at least one antigenic peptide or protein may be selected from any peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), and the RNA polymerase L (RAV-L) of any Rabies virus isolate or a fragment, variant or derivative thereof or from any synthetically engineered rabies peptide or protein.
  • RAV-G glycoprotein G
  • RAV-N nucleoprotein N
  • RAV-P the posphoprotein P
  • RV-M matrix protein M
  • RAV-L RNA polymerase L
  • the full-length protein of the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) is encoded by the coding region comprised in the inventive mRNA.
  • a fragment comprising at least one epitope of the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) is encoded by the coding region comprised in the inventive mRNA.
  • a Rabies vaccine strain preferably of the Pasteur vaccine strain according to the NCBI accession No. M13215: Glycoprotein G (RAV-G) of Pasteur vaccine strain:
  • RNA Polymerase L (RAV-L) of Pasteur vaccine strain
  • amino acid sequences of the Flury-LEP vaccine strain (used for Rabipur®) according to the NCBI accession No. GU565703:
  • RAV-G Glycoprotein G of Flury-LEP vaccine strain
  • amino acid sequences according to SEQ ID Nos. 1 -10 also amino acid sequences of different Rabies virus isolates can be used according to the invention and are incorporated herewith. These different Rabies virus isolates show preferably an identity of at least 70%, more preferably of at least 80% and most preferably of at least 90% with the amino acid sequences according to SEQ ID Nos. 1 -10.
  • Rabies virus strains according to the NCBI Accession Nos. JQ730682, AF499686, AB569299, AB8391 70, AB781935, FJ959397, AB362483, EF206720, EF20671 8, EF20671 7, EF206715, EF206714, EF206713, EF206712, EF20671 1 , EF206710, EF206709, EF206708, EF206707, EU182346, HM535790, GQ91 8139, EU877071 , EU877070, EU877069, EU1 82347, M31046, EU877068, EU877067, EF542830, AB8391 69, JQ647510, KC1 69986, JX088694, JQ730682, JN609295, JN23441 1 , HQ31 791 8, EF20671 9, EF5641 74, EU643590,
  • the coding region encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) of Rabies virus or a fragment, variant or derivative thereof may be selected from any nucleic acid sequence comprising a coding region derived from any Rabies virus isolate or a fragment or variant thereof.
  • a Rabies virus vaccine strain preferably of the Pasteur vaccine strain according to the NCBI accession No. ⁇ 3215.
  • Phosphoprotein P also named M1 (RAV-P) of Pasteur vaccine strain:
  • Matrix protein M also named M2 (RAV-M) of Pasteur vaccine strain:
  • nucleic acid sequences of different Rabies virus isolates are incorporated herewith. These different Rabies virus isolates show preferably an identity of at least 50%, 60%, 70%, more preferably of at least 80% and most preferably of at least 90% with the nucleic acid sequences according to SEQ ID Nos. 1 1 -1 5 or of fragments thereof.
  • Rabies virus strains according to the NCBI Accession Nos. JQ730682, AF499686, AB569299, AB8391 70, AB781935, FJ959397, AB362483, EF206720, EF20671 8, EF20671 7, EF20671 5, EF206714, EF206713, EF206712, EF20671 1 , EF206710, EF206709, EF206708, EF206707, EU182346, HM535790, GQ918139, EU877071 , EU877070, EU877069, EU1 82347, M31046, EU877068, EU877067, EF542830, AB8391 69, JQ647510, KC1 69986, JX088694, JQ730682, JN609295, JN23441 1 , HQ31 791 8, EF20671 9, EF5641 74, EU643590, J
  • the mRNA according to the invention does not comprise a reporter gene or a marker gene.
  • the mRNA according to the invention does not encode, for instance, luciferase; green fluorescent protein (GFP) and its variants (such as eGFP, RFP or BFP); a-globin; hypoxanthine-guanine phosphoribosyltransferase (HGPRT); ⁇ - galactosidase; galactokinase; alkaline phosphatase; secreted embryonic alkaline phosphatase (SEAP)) or a resistance gene (such as a resistance gene against neomycin, puromycin, hygromycin and zeocin).
  • the mRNA according to the invention does not encode luciferase.
  • the mRNA according to the invention does not encode GFP or a variant thereof.
  • the mRNA according to the invention does not encode a protein (or a fragment of a protein) derived from a virus belonging to the family of Orthomyxoviridae.
  • the mRNA does not encode a protein that is derived from an influenza virus, more preferably an influenza A virus.
  • the mRNA according to the invention does not encode an influenza A protein selected from the group consisting of hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), Ml , M2, NS1 , NS2 (NEP: nuclear export protein), PA, PB1 (polymerase basic 1 ), PB1 -F2 and PB2.
  • the mRNA according to the invention does not encode ovalbumin (OVA) or a fragment thereof.
  • the mRNA according to the invention does not encode an influenza A protein or ovalbumin.
  • the inventive mRNA preferably comprises at least one of the following structural elements: a 5'- and/or 3'- untranslated region element (UTR element), particularly a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene or from a fragment, homolog or a variant thereof, or a 5'- and/or 3'-UTR element which may be derivable from a gene that provides a stable mRNA or from a homolog, fragment or variant thereof; a histone-stem-loop structure, preferably a histone-stem-loop in its 3' untranslated region; a 5'-CAP structure; a poly-A tail ⁇ or a poly(C) sequence.
  • UTR element 5'- and/or 3'- untranslated region element
  • the inventive mRNA comprises at least one 5'- or 3'-UTR element.
  • an UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'- or 3'-UTR of any naturally occurring gene or which is derived from a fragment, a homolog or a variant of the 5'- or 3'- UTR of a gene.
  • the 5'- or 3'-UTR element used according to the present invention is heterologous to the coding region of the inventive mRNA sequence. Even if 5'- or 3'-UTR elements derived from naturally occurring genes are preferred, also synthetically engineered UTR elements may be used in the context of the present invention.
  • the inventive mRNA sequence comprises at least one 5'-untranslated region element (5'UTR element) which comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'UTR of a TOP gene.
  • 5'UTR element 5'-untranslated region element
  • the 5'UTR element does not comprise a TOP-motif or a 5'TOP, as defined above.
  • the nucleic acid sequence of the 5'UTR element which is derived from a 5'UTR of a TOP gene terminates at its 3'-end with a nucleotide located at position 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 upstream of the start codon (e.g. A(U T)G) of the gene or mRNA it is derived from.
  • the 5'UTR element does not comprise any part of the protein coding region.
  • the only protein coding part of the inventive mRNA is provided by the coding region.
  • the nucleic acid sequence which is derived from the 5'UTR of a TOP gene is derived from a eukaryotic TOP gene, preferably a plant or animal TOP gene, more preferably a chordate TOP gene, even more preferably a vertebrate TOP gene, most preferably a mammalian TOP gene, such as a human TOP gene.
  • the 5'UTR element is prefereably selected from 5'-UTR elements comprising or consisting of a nucleic acid sequence which is derived from a nucleic acid sequence selected from the group consisting of SEQ ID Nos. 1 -1363, SEQ ID NO. 1 395, SEQ ID NO.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a nucleic acid sequence extending from nucleotide position 5 (i.e. the nucleotide that is located at position 5 in the sequence) to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO201 3/143700, from the homologs of SEQ ID Nos.
  • the 5' UTR element is derived from a nucleic acid sequence extending from the nucleotide position immediately 3' to the 5'TOP to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal protein or from a variant of a 5'UTR of a TOP gene encoding a ribosomal protein.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 1 70, 193, 244, 259, 554, 650, 675, 700, 721 , 91 3, 101 6, 1063, 1 120, 1 138, and 1284-1 360 of the patent application WO201 3/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'TOP motif.
  • the sequence extending from position 5 to the nucleotide immediately 5' to the ATG corresponds to the 5'UTR of said sequences.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog or variant of a 5'UTR of a TOP gene encoding a ribosomal Large protein (RPL).
  • RPL ribosomal Large protein
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 259, 1284-131 8, 1344, 1346, 1 348-1354, 1357, 1358, 1421 and 1422of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'TOP motif.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, or from a variant of the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, wherein preferably the 5'UTR element does not comprise the 5'TOP of said gene.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1 6 (5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract:
  • the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No.
  • the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR.
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the inventive mRNA comprises a 5'UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a vertebrate TOP gene, such as a mammalian, e.g.
  • a human TOP gene selected from RPSA, RPS2, RPS3, RPS3A, RPS4, RPS5, RPS6, RPS7, RPS8, RPS9, RPS10, RPS1 1 , RPS12, RPS1 3, RPS14, RPS1 5, RPS1 5A, RPS16, RPS1 7, RPS1 8, RPS19, RPS20, RPS21 , RPS23, RPS24, RPS25, RPS26, RPS27, RPS27A, RPS28, RPS29, RPS30, RPL3, RPL4, RPL5, RPL6, RPL7, RPL7A, RPL8, RPL9, RPL10, RPL10A, RPL1 1 , RPL12, RPL1 3, RPL1 3A, RPL14, RPL1 5, RPL1 7, RPL1 8, RPL1 8A, RPL1 9, RPL21 , RPL22, RPL23, RPL23A, RPL24, RPL26, RPL27, RPL27A,
  • the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21 ), an ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 , cardiac muscle (ATP5A1 ) gene, an hydroxysteroid (1 7-beta) dehydrogenase 4 gene (HSD1 7B4), an androgen-induced 1 gene (AIG1 ), cytochrome c oxidase subunit Vic gene (COX6C), or a N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1 ) or from a variant thereof, preferably from a vertebrate ribosomal protein Large 32 gene (RPL32), a vertebrate ribosomal protein Large 32
  • the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No.
  • the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1368, or SEQ ID NOs 1412-1420 of the patent application WO2013/143700, wherein, preferably, the fragment is as described above, i.e.
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 26 (5'-UTR of ATP5A1 lacking the 5' terminal oligopyrimidine tract: GCGGCTCGGCCATTTTGTCCCAGTCAGTCCGGAGGCTGCGGCTGCAGAAGTACCGCCT GCG-GAGTAACTGCAAAG; corresponding to SEQ ID No.
  • the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 26 or more preferably to a corresponding RNA sequence, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc.
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the inventive mRNA further comprises at least one 3'UTR element which comprises or consists of a nucleic acid sequence derived from the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
  • '3'UTR element' refers to a nucleic acid sequence which comprises or consists of a nucleic acid sequence that is derived from a 3'UTR or from a variant of a 3'UTR.
  • a 3'UTR element in the sense of the present invention may represent the 3'UTR of an mRNA.
  • a 3'UTR element may be the 3'UTR of an mRNA, preferably of an artificial mRNA, or it may be the transcription template for a 3'UTR of an mRNA.
  • a 3'UTR element preferably is a nucleic acid sequence which corresponds to the 3'UTR of an mRNA, preferably to the 3'UTR of an artificial mRNA, such as an mRNA obtained by transcription of a genetically engineered vector construct.
  • the 3'UTR element fulfils the function of a 3'UTR or encodes a sequence which fulfils the function of a 3'UTR.
  • the inventive mRNA comprises a 3'UTR element which may be derivable from a gene that relates to an mRNA with an enhanced half-life (that provides a stable mRNA), for example a 3'UTR element as defined and described below.
  • the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1 (I) gene, or from a variant of a 3'UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1 (I) gene according to SEQ ID No.
  • the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an albumin gene, preferably a vertebrate albumin gene, more preferably a mammalian albumin gene, most preferably a human albumin gene according to SEQ ID No. 1 7
  • the inventive mRNA comprises a 3'-UTR element comprising a corresponding RNA sequence derived from the nucleic acids according to SEQ ID No. 1369-1390 of the patent application WO2013/143700 or a fragment, homolog or variant thereof.
  • the 3'-UTR element comprises the nucleic acid sequence derived from a fragment of the human albumin gene according to SEQ ID No. 18:
  • the 3'-UTR element of the inventive mRNA comprises or consists of a corresponding RNA sequence of the nucleic acid sequence according to SEQ ID No. 18.
  • the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an a-globin gene, preferably a vertebrate a-or ⁇ -globin gene, more preferably a mammalian cc-or ⁇ -globin gene, most preferably a human a-or ⁇ -globin gene according to SEQ ID No. 1 9-21 : 3'-UTR of Homo sapiens hemoglobin, alpha 1 (HBA1 )
  • HBA2 Homo sapiens hemoglobin, alpha 2
  • HBB Homo sapiens hemoglobin, beta
  • the 3'UTR element may comprise or consist of the center, a-complex-binding portion of the 3'UTR of an a-globin gene, such as of a human a-globin gene, preferably according to SEQ ID No. 22:
  • ⁇ -globin gene also named herein as "muag"
  • the 3'-UTR element of the inventive mRNA comprises or consists of a corresponding RNA sequence of the nucleic acid sequence according to SEQ ID No. 22 or a homolog, a fragment or variant thereof.
  • nucleic acid sequence which is derived from the 3'UTR of a noted gene' preferably refers to a nucleic acid sequence which is based on the 3'UTR sequence of a noted gene or on a part thereof, such as on the 3'UTR of an albumin gene, an ⁇ -globin gene, a ⁇ - globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1 (1) gene, preferably of an albumin gene or on a part thereof.
  • This term includes sequences corresponding to the entire 3'UTR sequence, i.e.
  • the full length 3'UTR sequence of a gene and sequences corresponding to a fragment of the 3'UTR sequence of a gene, such as an albumin gene, a-globin gene, ⁇ -globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1 (1) gene, preferably of an albumin gene.
  • a gene such as an albumin gene, a-globin gene, ⁇ -globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1 (1) gene, preferably of an albumin gene.
  • nucleic acid sequence which is derived from a variant of the 3'UTR of a noted gene' preferably refers to a nucleic acid sequence which is based on a variant of the 3'UTR sequence of a gene, such as on a variant of the 3'UTR of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1 (1) gene, or on a part thereof as described above.
  • This term includes sequences corresponding to the entire sequence of the variant of the 3'UTR of a gene, i.e.
  • a fragment in this context preferably consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length variant 3'UTR, which represents at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length variant 3'UTR.
  • Such a fragment of a variant in the sense of the present invention, is preferably a functional fragment of a variant as described herein.
  • the at least one 5'UTR element and the at least one 3'UTR element act synergistically to increase protein production from the inventive mRNA as described above.
  • the inventive mRNA comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises a histone stem-loop sequence/structure.
  • histone stem- loop sequences are preferably selected from histone stem-loop sequences as disclosed in WO 2012/01 9780, whose disclosure is incorporated herewith by reference.
  • a histone stem-loop sequence suitable to be used within the present invention, is preferably selected from at least one of the following formulae (I) or (II): formula (I) (stem-loop sequence without stem bordering elements):
  • N1-6 [N0-2GN3-5] [No-4(U/T)No- 4 ] [N3-5CN0-2] Ni-6 steml steml loop stem2 stem2 bordering element bordering element wherein: steml or stem2 bordering elements N1-6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof;
  • steml is reverse complementary or partial ly reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N 0 - 2 is a consecutive sequence of 0 to 2, preferably of 0 to 1, more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof, and wherein G is guanosine or an analogue thereof, and may be optionally replaced by a cytidine or an analogue thereof, provided that its complementary nucleotide cytidine in stem2 is replaced by guanosine; loop sequence [N 0 -4(U/T
  • U/T represents uridine, or optionally thymidine
  • stem2 [N3-5CN0-2] is reverse complementary or partially reverse complementary with element steml , and is a consecutive sequence between of 5 to 7 nucleotides
  • N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T,
  • N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G or C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleoside guanosine in stemi is replaced by cytidine; wherein
  • stemi and stem2 are capable of base pairing with each other forming a reverse complementary sequence, wherein base pairing may occur between stemi and stem2, e.g. by Watson-Crick base pairing of nucleotides A and U T or G and C or by non-Watson-Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between stemi and stem2, on the basis that one ore more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
  • the inventive mRNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (la) or (11a): formula (la) (stem-loop sequence without stem bordering elements):
  • stemi loop stem2 formula (I la) (stem-loop sequence with stem bordering elements):
  • N 2 -5 [N0-1 G N3-5] [Ni -3 (U/T)No-2] [ N3-5CN0-1 ] N2-5
  • bordering element bordering element wherein: N, C, G, T and U are as defined above.
  • the inventive mRNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (lb) or (Mb): formula (lb) (stem-loop sequence without stem bordering elements):
  • stem 1 loop stem2 formula (lib) (stem-loop sequence with stem bordering elements): N4-5 [Ni GNJ [N 2 (U T)Ni ] [N4CN1] N4-5 steml steml loop stem2 stem2
  • a particular preferred histone stem-loop sequence is the sequence according to SEQ ID NO: 23 CAAAGGCTCTTTTCAGAGCCACCA or more preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 27 (CAAAGGCUCUUUUCAGAGCCACCA SEQ ID NO: 27).
  • the inventive mRNA comprises additionally to the coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, a poly(A) sequence, also called poly-A-tail, preferably at the 3'-terminus of the inventive mRNA.
  • such a poly(A) sequence comprises a sequence of about 25 to about 400 adenosine nucleotides, preferably a sequence of about 50 to about 400 adenosine nucleotides, more preferably a sequence of about 50 to about 300 adenosine nucleotides, even more preferably a sequence of about 50 to about 250 adenosine nucleotides, most preferably a sequence of about 60 to about 250 adenosine nucleotides.
  • the term "about” refers to a deviation of ⁇ 10% of the value(s) it is attached to.
  • This poly(A) sequence is preferably located 3' of the coding region comprised in the inventive mRNA according to the first aspect of the present invention.
  • the inventive mRNA can be modified by a sequence of at least 1 0 cytosines, preferably at least 20 cytosines, more preferably at least 30 cytosines (so-called "poly(C) sequence").
  • the mRNA may contain a poly(C) sequence of typically about 1 0 to 200 cytosine nucleotides, preferably about 10 to 100 cytosine nucleotides, more preferably about 1 0 to 70 cytosine nucleotides or even more preferably about 20 to 50 or even 20 to 30 cytosine nucleotides.
  • This poly(C) sequence is preferably located 3' of the coding region, more preferably 3' of an optional poly(A) sequence comprised in the inventive mRNA according to the first aspect of the present invention.
  • the inventive mRNA sequence may comprise in a specific embodiment: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c.) a poly(A) sequence preferably comprising 64 adenosines; and d.) optionally, a poly(C) sequence, preferably comprising 30 cytosines.
  • the inventive mRNA comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c. ) a poly(A) sequence preferably comprising 64 adenosines; d. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and e. ) a histone-stem-loop, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 23.
  • the inventive mRNA comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c. ) optionally, a 3'-UTR element derived from an alpha globin gene, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO.
  • the inventive mRNA encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b.
  • a 5'-UTR element derived from a TOP gene preferably derived from the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6 , a homolog, a fragment, or a variant thereof; c. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; d. ) optionally, a 3'UTR element derived of a gene providing a stable mRNA, preferably derived from the corresponding RNA sequence of a nucleic acid sequence according to SEQ ID NO. 18, a homolog, a fragment, or a variant thereof; e.
  • a poly(A) sequence preferably comprising 64 adenosines
  • a poly(C) sequence preferably comprising 30 cytosines
  • a histone-stem-loop preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 23.
  • the coding region might encode at least partially one of the amino acid sequences according to SEQ ID Nos. 1 -10 or fragments, variants or derivatives thereof. Furthermore the coding region of the inventive mRNA may encode a combination of at least two of these amino acid sequences or a combination of fragments, variants or derivatives thereof.
  • the coding region might be or might comprise at least partially one of the sequences according to SEQ ID No. 1 1 to SEQ ID No. 1 5, or fragments, homologs or variants thereof.
  • the mRNA might comprise a combination of at least two of these sequences or a combination of fragments, homologs or variants thereof.
  • the inventive mRNA may be provided as a stabilized nucleic acid, e.g. in the form of a modified nucleic acid.
  • the inventive mRNA is stabilized, preferably by backbone modifications, sugar modifications and/or base modifications, more preferred stabilized by modification of the G/C-content. All of these modifications may be introduced into the inventive mRNA without impairing the mRNA's function to be translated into the antigenic function derived from the Rabies virus peptide or protein.
  • a backbone modification in the context of the present invention is preferably a modification in which phosphates of the backbone of the nucleotides contained in the inventive mRNA are chemically modified, e.g. anionic i nternucleoside li nkage, N3'->P5' modifications, replacement of non-bridging oxygen atoms by boranes, neutral internucleoside linkage, amide linkage of the nucleosides, methylene(methylimino) linkages, formacetal and thioformacetal linkages, introduction of sulfonyl groups, or the like.
  • a sugar modification in the context of the present invention is preferably a chemical modification of the sugar of the nucleotides of the inventive mRNA, e.g. methylation of the ribose residue or the like.
  • the inventive mRNA may be modified and thus stabilized by modifying the G (guanosine)/C (cytosine) content of the mRNA, preferably of the coding region thereof.
  • the G/C content of the inventive mRNA is particularly increased compared to the G/C content of the coding region of its particular wild type coding sequence, i.e. the unmodified mRNA.
  • the encoded amino acid sequence of the inventive mRNA is preferably not modified compared to the coded amino acid sequence of the particular wild type/unmodified mRNA.
  • the modification of the G/C-content of the inventive mRNA is based on the fact that RNA sequences having an increased G (guanosine)/C (cytosine) content are more stable than RNA sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of a coding sequence or a whole RNA might therefore be varied compared to the wild type coding sequence or mRNA, such that they include an increased amount of G/C nucleotides whi le the translated amino acid sequence is retained.
  • the most favourable codons for the stabi lity can be determined (so-called alternative codon usage).
  • the G/C content of the coding region of the inventive mRNA according to the invention is increased by at least 7%, more preferably by at least 1 5%, particularly preferably by at least 20%, compared to the G/C content of the coded region of the wi ld type RNA.
  • at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, more preferably at least 70 %, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for a protein or peptide as defined herein or its fragment or variant thereof or the whole sequence of the wild type mRNA sequence or coding sequence are substituted, thereby increasing the G/C content of said sequence.
  • the inventive mRNA is optimized for translation, preferably optimized for translation by replacing codons for less frequent tRNAs of a given amino acid by codons for more frequently occurring tRNAs of the respective amino acid. This is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, if so-called "less frequent codons" are present in the inventive mRNA to an increased extent, the corresponding modified RNA is translated to a significantly poorer degree than in the case where codons coding for more frequent tRNAs are present.
  • the coding region of the inventive mRNA is modified compared to the corresponding region of the wild type RNA or coding sequence such that at least one codon of the wild type sequence which codes for a tRNA which is relatively rare or less frequent in the cell is exchanged for a codon which codes for a tRNA which is more or most frequent in the cell and carries the same amino acid as the relatively rare or less frequent tRNA.
  • the sequences of the inventive mRNA can be modified such that codons for which more frequently occurring tRNAs are available are inserted.
  • Substitutions, additions or eliminations of bases are preferably carried out using a DNA matrix for preparation of the nucleic acid molecule by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation.
  • a corresponding DNA molecule may be transcribed in vitro.
  • This DNA matrix preferably comprises a suitable promoter, e.g. a T7 or SP6 promoter, for in vitro transcription, which is followed by the desired nucleotide sequence for the at least one RNA to be prepared and a termination signal for in vitro transcription.
  • the DNA molecule which forms the matrix of the at least one RNA of interest, may be prepared by fermentative proliferation and subsequent isolation as part of a plasmid which can be replicated in bacteria.
  • Plasmids which may be mentioned as suitable for the present invention are e.g. the plasmids pT7Ts (GenBank accession number U26404; Lai et a/., Development 1995, 121 : 2349 to 2360), pGEM ® series, e.g. pGEM ® -1 (GenBank accession number X65300; from Promega) and pSP64 (GenBank accession number X65327); cf.
  • the inventive mRNA sequence according to the first aspect of the present invention comprises, preferably in 5'- to 3'- direction:
  • a) a 5'-CAP structure as defined herein, preferably m7GpppN;
  • a coding region preferably with an increased or even maximized G/C content compared with the G/C content of the coding region of the wild type mRNA, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof; c) a 3'-UTR element as defined herein, preferably derived of a gene providing a stable mRNA, most preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22, or a homolog, a fragment or variant thereof;
  • a poly(A) sequence preferably consisting of 64 adenosines
  • e) optionally a poly(C) sequence, preferably consisting of 30 cytosines.
  • At least one histone stem-loop sequence preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 23.
  • the inventive mRNA sequence of that specific embodiment comprises the sequence modifications as shown in Fig. 1 (SEQ ID NO. 24).
  • the inventive mRNA sequence according to the first aspect of the present invention comprises preferably in 5' to 3' direction: a) a 5'-CAP structure, as defined herein, preferably m7GpppN;
  • a 5'-UTR element as defined herein, preferably a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, preferably the 5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract according to SEQ ID NO. 1 6 or the corresponding RNA sequence; or a fragment, homolog or variant thereof;
  • a coding region preferably with an increased or even maximized G/C content compared with the G/C content of the coding region of the wild type mRNA, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), and the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof;
  • a 3'-UTR element preferably the 3'-UTR element of human albumin according to SEQ ID No. 18 or the corresponding RNA, or a homolog, a fragment or a variant thereof; e) a poly(A) sequence, preferably consisting of 64 adenosines
  • f) optionally a poly(C) sequence, preferably consisting of 30 cytosines.
  • the inventive mRNA of that specific embodiment comprises the sequence modifications as shown in Fig. 2 (SEQ ID NO. 25).
  • inventive mRNA comprises or consists of the sequences shown in Fig. 1 or 2 according to SEQ ID Nos. 24 and 25.
  • the mRNA according to the invention may further comprise an internal ribosome entry site (IRES) sequence or IRES-motif, which may separate several open reading frames, for example if the inventive mRNA encodes for two or more antigenic peptides or proteins.
  • IRES-sequence may be particularly helpful if the mRNA is a bi- or multicistronic mRNA.
  • the inventive mRNA may be prepared using any method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as in vitro methods, such as in vitro transcription reactions.
  • the mRNA comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof may be administered naked without being associated with any further vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the inventive mRNA or of further comprised nucleic acid.
  • the inventive mRNA may be formulated together with a cationic or polycationic compound and/or with a polymeric carrier. Accordingly, in a further embodiment of the invention it is preferred that the inventive mRNA or any other nucleic acid comprised in the inventive pharmaceutical composition or vaccine is associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w) of mRNA or nucleic acid to cationic or polycationic compound and/or with a polymeric carrier; or optionally in a
  • inventive mRNA or any other nucleic acid comprised in the inventive pharmaceutical composition or vaccine can also be associated with a vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the inventive mRNA or of optionally comprised further included nucleic acids.
  • Cationic or polycationic compounds being particularly preferred agents in this context include protamine, nucleoline, spermine or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs), PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1 , L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, plsl,
  • protamine is particularly preferred.
  • preferred cationic or polycationic proteins or peptides may be selected from the following proteins or peptides having the following total formula (III):
  • Particularly preferred cationic peptides in this context are e.g. Argy, Arg 8 , Arg 9 , H 3 R 9 , R 9 H 3 , H 3 R 9 H 3 , YSSR 9 SSY, (RKH) 4 , Y(RKH) 2 R, etc.
  • cationic or polycationic compounds which can be used as transfection or complexation agent may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • cationic polysaccharides for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • PEI polyethyleneimine
  • DOTMA [1 -(2,3- sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Choi, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol- amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DIMRI: Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3- (trimethylammonio)propane, DC-6-14: O,O-ditetradecanoyl-N-(a- trimethylammonioacetyl)diethanolamine chloride, CLIP1 : rac-[(2,3-dioctadecyloxypropyl)(2- hydroxyethyl)]-d
  • modified polyaminoacids such as ⁇ -aminoacid-polymers or reversed polyamides, etc.
  • modified polyethylenes such as PVP (poly(N-ethyl-4-vinylpyridinium bromide)), etc.
  • modified acrylates such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc.
  • modified amidoamines such as pAMAM (poly(amidoamine)), etc.
  • modified polybetaaminoester (PBAE) such as diamine end modified 1 ,4 butanediol diacrylate-co-5-amino-1 -pentanol polymers, etc.
  • dendrimers such as polypropylamine dendrimers or pAMAM based dendrimers, etc.
  • polyimine(s) such as PEI: poly(ethyleneimine), poly(propyleneimine), etc.
  • polyallylamine sugar backbone based
  • a polymeric carrier used according to the invention might be a polymeric carrier formed by disulfide-crosslinked cationic components.
  • the disulfide-crosslinked cationic components may be the same or different from each other.
  • the polymeric carrier can also contain further components.
  • the polymeric carrier used according to the present invention comprises mixtures of cationic peptides, proteins or polymers and optionally further components as defined herein, which are crosslinked by disulfide bonds as described herein.
  • the disclosure of WO 2012/013326 is incorporated herewith by reference.
  • the cationic components which form basis for the polymeric carrier by disulfide-crosslinkage, are typically selected from any suitable cationic or polycationic peptide, protein or polymer suitable for this purpose, particular any cationic or polycationic peptide, protein or polymer capable to complex an mRNA or a nucleic acid as defined according to the present invention, and thereby preferably condensing the mRNA or the nucleic acid.
  • the cationic or polycationic peptide, protein or polymer is preferably a linear molecule, however, branched cationic or polycationic peptides, proteins or polymers may also be used.
  • Every disulfide-crosslinking cationic or polycationic protein, peptide or polymer of the polymeric carrier which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine contains at least one -SH moiety, most preferably at least one cysteine residue or any further chemical group exhibiting an -SH moiety, capable to form a disulfide linkage upon condensation with at least one further cationic or polycationic protein, peptide or polymer as cationic component of the polymeric carrier as mentioned herein.
  • the polymeric carrier which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine may be formed by disulfide-crosslinked cationic (or polycationic) components.
  • such cationic or polycationic peptides or proteins or polymers of the polymeric carrier which comprise or are additionally modified to comprise at least one -SH moiety, are selected from, proteins, peptides and polymers as defined above for complexation agent.
  • the polymeric carrier which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine may be selected from a polymeric carrier molecule according to generic formula (IV):
  • P 1 and P 3 are different or identical to each other and represent a linear or branched hydrophilic polymer chain, each P' and P 3 exhibiting at least one -SH-moiety, capable to form a disulfide linkage upon condensation with component P 2 , or alternatively with (AA), (AA) X , or [(AA) X ] 2 if such components are used as a linker between P 1 and P 2 or P 3 and P 2 ) and/or with further components (e.g.
  • hydrophilic polymer chain exhibits a molecular weight of about 1 kDa to about 100 kDa, preferably of about 2 kDa to about 25 kDa; or more preferably of about 2 kDa to about 1 0 kDa, e.g. about 5 kDa to about 25 kDa or 5 kDa to about 10 kDa;
  • P 2 is a cationic or polycationic peptide or protein, e.g. as defined above for the polymeric carrier formed by disulfide-crosslinked cationic components, and preferably having a length of about 3 to about 1 00 amino acids, more preferably having a length of about 3 to about 50 amino acids, even more preferably having a length of about 3 to about 25 amino acids, e.g. a length of about 3 to 10, 5 to 1 5, 10 to 20 or 1 5 to 25 amino acids, more preferably a length of about 5 to about 20 and even more preferably a length of about 1 0 to about 20; or is a cationic or polycationic polymer, e.g.
  • the polymeric carrier formed by disulfide-crosslinked cationic components typically having a molecular weight of about 0.5 kDa to about 30 kDa, including a molecular weight of about 1 kDa to about 20 kDa, even more preferably of about 1 .5 kDa to about 10 kDa, or having a molecular weight of about 0.5 kDa to about 100 kDa, including a molecular weight of about 10 kDa to about 50 kDa, even more preferably of about 10 kDa to about 30 kDa; each P 2 exhibiting at least two -SH-moieties, capable to form a disulfide linkage upon condensation with further components P 2 or component(s) P 1 and/or P 3 or alternatively with further components (e.g.
  • (AA), (AA) X , or [(AA) X ] Z ); is a (reversible) disulfide bond (the brackets are omitted for better readability), wherein S preferably represents sulphur or a -SH carrying moiety, which has formed a (reversible) disulfide bond.
  • the (reversible) disulfide bond is preferably formed by condensation of -SH-moieties of either components P 1 and P 2 , P 2 and P 2 , or P 2 and P 3 , or optionally of further components as defined herein (e.g.
  • the -SH-moiety may be part of the structure of these components or added by a modification as defined below; is an optional ligand, which may be present or not, and may be selected independent from the other from RGD, Transferrin, Folate, a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide, (e.g. TAT or KALA), a ligand of a receptor (e.g. cytokines, hormones, growth factors etc), small molecules (e.g.
  • n is in a range of about 1 , 2, 3, 4, or 5 to 10, more preferably in a range of about 1 , 2, 3, or 4 to 9, in a range of about 1 , 2, 3, or 4 to 8, or i n a range of about 1 , 2, or 3 to 7.
  • Each of hydrophilic polymers P 1 and P 3 typical ly exhibits at least one -SH-moiety, wherein the at least one -SH-moiety is capable to form a disulfide linkage upon reaction with component P 2 or with component (AA) or (AA) X , if used as linker between P 1 and P 2 or P 3 and P 2 as defined below and optionally with a further component, e.g. L and/or (AA) or (AA) X , e.g. if two or more -SH-moieties are contained.
  • -SH-moieties are typically provided by each of the hydrophilic polymers P 1 and P 3 , e.g. via an internal cysteine or any further (modified) amino acid or compound which carries a -SH moiety.
  • the subformulae "P 1 -S-S-P 2 " and “P 2 -S-S-P 3 " may also be written as "P'-Cys- Cys-P 2 " and "P 2 -Cys-Cys-P 3 ", if the -SH- moiety is provided by a cysteine, wherein the term Cys-Cys represents two cysteines coupled via a disulfide bond, not via a peptide bond.
  • the term "-S-S-" in these formulae may also be written as “-S-Cys", as “-Cys-S” or as “-Cys-Cys-”.
  • the term “-Cys-Cys-” does not represent a peptide bond but a linkage of two cysteines via their -SH-moieties to form a disulfide bond.
  • the term “-Cys-Cys-” also may be understood generally as “-(Cys-S)-(S-Cys)-", wherein in this specific case S indicates the sulphur of the -SH-moiety of cysteine.
  • the terms "-S- Cys” and “-Cys-S” indicate a disulfide bond between a -SH containing moiety and a cysteine, which may also be written as “-S-(S-Cys)" and "-(Cys-S)-S".
  • the hydrophilic polymers P 1 and P 3 may be modified with a -SH moiety, preferably via a chemical reaction with a compound carrying a -SH moiety, such that each of the hydrophilic polymers P 1 and P 3 carries at least one such -SH moiety.
  • a compound carrying a -SH moiety may be e.g.
  • Such a compound may also be any non-amino compound or moiety, which contains or al lows to introduce a -SH moiety into hydrophilic polymers P 1 and P 3 as defined herein.
  • Such non-amino compounds may be attached to the hydrophilic polymers P 1 and P 3 of formula (VI) of the polymeric carrier according to the present invention via chemical reactions or binding of compounds, e.g. by binding of a 3-thio propionic acid or thioimolane, by amide formation (e.g.
  • alkenes or alkines imine or hydrozone formation (aldehydes or ketons, hydrazins, hydroxylamins, amines), complexation reactions (avidin, biotin, protein G) or components which allow S n - type substitution reactions (e.g halogenalkans, thiols, alcohols, amines, hydrazines, hydrazides, sulphonic acid esters, oxyphosphonium salts) or other chemical moieties which can be utilized in the attachment of further components.
  • a particularly preferred PEG derivate in this context is alpha-Methoxy-omega-mercapto poly(ethylene glycol).
  • the SH-moiety e.g.
  • each of hydrophilic polymers P 1 and P 3 typically exhibits at least one -SH-moiety preferably at one terminal end, but may also contain two or even more - SH-moieties, which may be used to additionally attach further components as defined herein, preferably further functional peptides or proteins e.g. a ligand, an amino acid component (AA) or (AA) X , antibodies, cell penetrating peptides or enhancer peptides (e.g. TAT, KALA), etc.
  • further functional peptides or proteins e.g. a ligand, an amino acid component (AA) or (AA) X , antibodies, cell penetrating peptides or enhancer peptides (e.g. TAT, KALA), etc.
  • inventive mRNA is complexed at least partially with a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides.
  • a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides.
  • WO 2010/037539 and WO 2012/1 1351 3 is incorporated herewith by reference. Partially means that only a part of the inventive mRNA is complexed with a cationic compound and that the rest of the inventive mRNA is (comprised in the inventive pharmaceutical compostion or vaccine) in uncomplexed form ("free").
  • the ratio of complexed mRNA to: free mRNA is selected from a range of about 5:1 (w/w) to about 1 :10 (w/w), more preferably from a range of about 4:1 (w/w) to about 1 :8 (w/w), even more preferably from a range of about 3:1 (w/w) to about 1 :5 (w/w) or 1 :3 (w/w), and most preferably the ratio of complexed mRNA to free mRNA in the inventive pharmaceutical composition or vaccine is selected from a ratio of about 1 :1 (w/w).
  • the complexed mRNA in the inventive pharmaceutical composition or vaccine is preferably prepared according to a first step by complexing the inventive mRNA with a cationic or polycationic compound and/or with a polymeric carrier, preferably as defined herein, in a specific ratio to form a stable complex.
  • a cationic or polycationic compound or polymeric carrier preferably as defined herein, in a specific ratio to form a stable complex.
  • the ratio of the mRNA and the cationic or polycationic compound and/or the polymeric carrier in the component of the complexed mRNA is typically selected in a range that the mRNA is entirely complexed and no free cationic or polycationic compound or polymeric carrier or only a negligibly small amount thereof remains in the composition.
  • the ratio of the mRNA to the cationic or polycationic compound and/or the polymeric carrier is selected from a range of about 6:1 (w/w) to about 0,25:1 (w/w), more preferably from about 5:1 (w/w) to about 0,5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w).
  • the ratio of the mRNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, in the component of the complexed mRNA may also be calculated on the basis of the nitrogen/phosphate ratio (N/P-ratio) of the entire complex.
  • an N/P-ratio is preferably in the range of about 0.1 -1 0, preferably in a range of about 0.3-4 and most preferably in a range of about 0.5-2 or 0.7-2 regarding the ratio of mRNA: cationic or polycationic compound and/or polymeric carrier, preferably as defined herein, in the complex, and most preferably in a range of about 0.7- 1 ,5, 0.5-1 or 0.7-1 , and even most preferably in a range of about 0.3-0.9 or 0.5-0.9., preferably provided that the cationic or polycationic compound in the complex is a cationic or polycationic cationic or polycationic protein or peptide and/or the polymeric carrier as defined above.
  • the complexed mRNA is also emcompassed in the term "adjuvant component".
  • the invention provides for a composition comprising a plurality or more than one, preferably 2 to 10, more preferably 2 to 5, most preferably 2 to 4 of the inventive mRNA sequences as defined herein.
  • inventive compositions comprise more than one inventive mRNA sequences, preferably encoding different peptides or proteins which comprise preferably different pathogenic antigens or fragments, variants or derivatives thereof.
  • at least one mRNA sequence encodes at least one antigenic peptide or protein derived from glycoprotein G (RAV-G) of Rabies virus and that at least one mRNA sequence encodes at least one antigenic peptide or protein derived from another antigen of Rabies virus, particularly of nucleoprotein N (RAV-N).
  • RAV-G glycoprotein G
  • RAV-N nucleoprotein N
  • the present invention also provides a pharmaceutical composition, comprising at least one inventive mRNA sequence as defined herein or an inventive composition comprising a plurality of inventive mRNA sequences as defined herein and optional ly a pharmaceutically acceptable carrier and/or vehicle.
  • inventive pharmaceutical composition comprises at least one inventive mRNA sequence as defined herein.
  • inventive pharmaceutical composition may optional comprise at least one additional pharmaceutical ly active component.
  • a pharmaceutically active component in this connection is a compound that has a therapeutic effect to heal, ameliorate or prevent a particular indication or disease as mentioned herein, preferably rabies.
  • Such compounds include, without implying any limitation, peptides or proteins, preferably as defined herein, nucleic acids, preferably as defi ned herein, (therapeutically active) low molecular weight organic or inorganic compounds (molecular weight less than 5000, preferably less than 1000), sugars, antigens or antibodies, preferably as defined herein, therapeutic agents already known in the prior art, antigenic cells, antigenic cel lular fragments, cel lular fractions; cell wall components (e.g.
  • rabies vaccines e.g. "Tol lwutimpfstoff (HDC) ineptiert” or Rabipur, or rabies immune globulines, e.g. Merieux P or Berirab.
  • the inventive pharmaceutical composition may be administered orally, parenteral ly, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques. Particularly preferred is intradermal and intramuscular injection.
  • Sterile injectable forms of the inventive pharmaceutical compositions may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the inventive vaccine may be administered by conventional needle injection or needle-free jet injection.
  • the inventive pharmaceutical composition may be administered by jet injection as defined herein, preferably intramuscularly or intradermal ly, more preferably intradermally.
  • the inventive pharmaceutical composition may comprise an adjuvant.
  • an adjuvant may be understood as any compound, which is suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response.
  • the inventive pharmaceutical composition when administered, preferably elicits an innate immune response due to the adjuvant, optionally contained therein.
  • such an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal, e.g. an adjuvant protein as defined above or an adjuvant as defined in the following.
  • adjuvants suitable for depot and delivery are cationic or polycationic compounds as defined above for the inventive mRNA sequence as vehicle, transfection or complexation agent.
  • the inventive pharmaceutical composition may comprise one or more additional adjuvants which are suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response, particularly by binding to pathogen-associated molecular patterns (PAMPs).
  • PAMPs pathogen-associated molecular patterns
  • the pharmaceutical composition or vaccine when administered, preferably elicits an innate immune response due to the adjuvant, optionally contained therein.
  • an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal, e.g. an adjuvant protein as defined above or an adjuvant as defined in the following.
  • such an adjuvant may be selected from an adjuvant as defined above.
  • an adjuvant may be selected from any adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal and/or suitable for depot and delivery of the components of the inventive pharmaceutical composition or vaccine.
  • adjuvants suitable for depot and delivery are cationic or polycationic compounds as defined above.
  • the adjuvant may be selected from the group consisting of, e.g., cationic or polycationic compounds as defined above, from chitosan, TDM, MDP, muramyl dipeptide, pluronics, alum solution, aluminium hydroxide, ADJUMERTM (polyphosphazene); aluminium phosphate gel; glucans from algae; algammulin; aluminium hydroxide gel (alum); highly protein-adsorbing aluminium hydroxide gel; low viscosity aluminium hydroxide gel; AF or SPT (emulsion of squalane (5%), Tween 80 (0.2%), Pluronic L121 (1 .25%), phosphate-buffered saline, pH 7.4); AVRIDINETM (propanediamine); BAY R1005TM ((N-(2-deoxy-2-L-leucylaminob- D- glucopyranosyl)-N-octadecyl-d
  • DMPC dehydroepiandrosterone
  • DMPG dimethyldioctadecylammonium chloride
  • ZnPro-8 zinc-L-proline salt complex
  • GMDP N-acetylglucosaminyl-(b1 -4)-N-acetylmuramyl-L47 alanyl-D- isoglutamine
  • TM liposomes
  • LOXORIBINETM (7-allyl-8-oxoguanosine); LT 5 oral adjuvant ⁇ E.coli labile enterotoxin-protoxin); microspheres and microparticles of any composition
  • MF59TM serum emulsion
  • MONTANIDE ISA 51 TM purified incomplete Freund's adjuvant
  • MONTANIDE ISA 720TM metabolisable oil adjuvant
  • MPLTM (3-Q-desacyl-4'-monophosphoryl lipid A
  • MTP-PE and MTP-PE liposomes ((N- acetyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1 ,2-dipalmitoyl-sn-glycero-3- (hydroxyphosphoryloxy))-ethylamide, monosodium salt);
  • MURAMETIDETM Nac-Mur-L-Ala- D-Gln-OC
  • an adjuvant may be selected from adjuvants, which support induction of a Th1 -immune response or maturation of naive T-cells, such as GM-CSF, IL-1 2, IFNg, any immunostimulatory nucleic acid as defined above, preferably an immunostimulatory RNA, CpG DNA, etc.
  • adjuvants which support induction of a Th1 -immune response or maturation of naive T-cells, such as GM-CSF, IL-1 2, IFNg, any immunostimulatory nucleic acid as defined above, preferably an immunostimulatory RNA, CpG DNA, etc.
  • the inventive pharmaceutical composition contains besides the antigen-providing mRNA further components which are selected from the group comprising: further antigens or further antigen-providing nucleic acids; a further immunotherapeutic agent; one or more auxiliary substances; or any further compound, which is known to be immunostimulating due to its binding affinity (as l igands) to human Toll-like receptors; and/or an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA).
  • further components which are selected from the group comprising: further antigens or further antigen-providing nucleic acids; a further immunotherapeutic agent; one or more auxiliary substances; or any further compound, which is known to be immunostimulating due to its binding affinity (as l igands) to human Toll-like receptors; and/or an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA).
  • the inventive pharmaceutical composition can additionally contain one or more auxiliary substances in order to increase its immunogenicity or immunostimulatory capacity, if desired.
  • a synergistic action of the inventive mRNA sequence as defined herein and of an auxiliary substance, which may be optionally contained in the inventive pharmaceutical composition, is preferably achieved thereby.
  • auxi liary substances various mechanisms can come into consideration in this respect.
  • compounds that permit the maturation of dendritic cells (DCs), for example lipopolysaccharides, TNF-alpha or CD40 ligand form a first class of suitable auxiliary substances.
  • auxiliary substance any agent that influences the immune system in the manner of a "danger signal” (LPS, GP96, etc.) or cytokines, such as GM-CFS, which allow an immune response to be enhanced and/or influenced in a targeted manner.
  • a "danger signal” LPS, GP96, etc.
  • cytokines such as GM-CFS
  • auxiliary substances are cytokines, such as monokines, lymphokines, interleukins or chemokines, that further promote the innate immune response, such as IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-1 3, IL-14, IL-1 5, IL-1 6, IL-1 7, IL-1 8, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta or TNF-alpha, growth factors, such as hGH.
  • cytokines such as monokines, lymphokines, inter
  • emulsifiers such as, for example, Tween ® ; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet- forming agents; stabilizers; antioxidants; preservatives.
  • the inventive pharmaceutical composition can also additionally contain any further compound, which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR1 3.
  • any further compound which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR1 3.
  • the optionally comprised adjuvant component comprises the same inventive m NA as comprised in the inventive pharmaceutical composition as antigen-providing mRNA e.g. mRNA coding for an antigenic peptide or protein of Rabies virus or fragments, variants or derivatives thereof.
  • inventive pharmaceutical composition may comprise further components for facilitating administration and uptake of components of the pharmaceutical composition.
  • Such further components may be an appropriate carrier or vehicle, additional adjuvants for supporting any immune response, antibacterial and/or antiviral agents.
  • the inventive pharmaceutical composition furthermore comprises a pharmaceutically acceptable carrier and/or vehicle.
  • Such a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of a composition comprising the components of the inventive pharmaceutical composition.
  • the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate etc. buffered solutions.
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e. the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g.
  • liquids occurring in "in vivd' methods such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitrd' methods, such as common buffers or liquids.
  • common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
  • compatible solid or liquid fillers or diluents or encapsulating compounds which are suitable for administration to a patient to be treated, may be used as well for the pharmaceutical composition according to the invention.
  • the term "compatible” as used here means that these constituents of the inventive pharmaceutical composition are capable of being mixed with the components of the inventive pharmaceutical composition in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the pharmaceutical compostion under typical use conditions.
  • a further component of the inventive pharmaceutical composition may be an immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably immunoglobulins directed against a Rabies virus, eg. Merieux P or Berirab.
  • such a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA.
  • a further immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive antigen-providing mRNA.
  • antigens can be included in the inventive pharmaceutical composition and are typically substances such as cells, cell lysates, viruses, attenuated viruses, inactivated viruses, proteins, peptides, nucleic acids or other bio- or macromolecules or fragments thereof.
  • antigens may be proteins and peptides or fragments thereof, such as epitopes of those proteins or peptides, preferably having 5 to 15, more preferably 6 to 9, amino acids.
  • said proteins, peptides or epitopes may be derived from Glycoprotein G(RAV- G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus or from fragments, variants or derivatives thereof.
  • antigens may also comprise any other biomolecule, e.g., lipids, carbohydrates, etc.
  • the antigen is a protein or (poly-) peptide antigen, a nucleic acid, a nucleic acid encoding a protein or (poly-) peptide antigen, a polysaccharide antigen, a polysaccharide conjugate antigen, a lipid antigen, a glycolipid antigen, a carbohydrate antigen, a bacterium, a cell (vaccine), or killed or attenuated viruses.
  • rabies vaccines comprising inactivated virus, as e.g. Rabipur comprising inactived Rabies virus strain Flury-LEP or "inattentionierter (HDC) Tollwutimpfstoff" comprising inactivated Rabies virus strain WISTAR PM/WI 38-1503-3M.
  • inventive pharmaceutical composition or vaccine as defined herein may furthermore comprise further additives or additional compounds.
  • Further additives which may be included in the pharmaceutical composition are emulsifiers, such as, for example, Tween ® ; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet-forming agents; stabilizers; antioxidants; preservatives, RNase inhibitors and/or an anti-bacterial agent or an anti-viral agent.
  • inventive pharmaceutical composition may comprise small interfering RNA (siRNA) directed against genes of Rabies virus, e.g.
  • siRNA directed against the gene encoding Glycoprotein G (RAV-G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus.
  • RAV-G Glycoprotein G
  • RAV-N nucleoprotein N
  • RAV-P phosphoprotein P
  • RAV-M matrix protein
  • RAV-L RNA polymerase L
  • the inventive pharmaceutical composition typically comprises a "safe and effective amount" of the components of the inventive pharmaceutical composition, particularly of the inventive mRNA sequence(s) as defined herein.
  • a "safe and effective amount” means an amount of the inventive mRNA sequence(s) as defined herein as such that is sufficient to significantly induce a positive modification of a disease or disorder or to prevent a disease, preferably rabies as defined herein.
  • a "safe and effective amount” is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • the inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
  • the inventive pharmaceutical composition (or the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein) may be provided or used as a vaccine.
  • a vaccine is as defined above for pharmaceutical compositions.
  • a vaccine typically contains the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein.
  • the inventive vaccine may also comprise a pharmaceutically acceptable carrier, adjuvant, and/or vehicle as defined herein for the inventive pharmaceutical composition.
  • the choice of a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered.
  • the inventive vaccine can be administered, for example, systemically or locally.
  • Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes.
  • Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections.
  • vaccines may be administered by an intradermal, subcutaneous, or intramuscular route.
  • inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form.
  • the inventive vaccine may be administered by conventional needle injection or needle-free jet injection.
  • the inventive vaccine may be administered by jet injection as defined herein, preferably intramuscularly or intradermal ly, more preferably intradermal ly.
  • the inventive vaccine can additionally contain one or more auxiliary substances in order to increase its immunogenicity or immunostimulatory capacity, if desired.
  • auxiliary substances particularly preferred are adjuvants as auxiliary substances or additives as defined for the pharmaceutical composition.
  • the invention is directed to a kit or kit of parts comprising the components of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine and optionally technical instructions with information on the administration and dosage of the components.
  • the kit may additionally contain a pharmaceutically acceptable vehicle, an adjuvant and at least one further component as defined herein, as wel l as means for administration and technical instructions.
  • the components of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine and e.g. the adjuvant may be provided in lyophilized form.
  • the provided vehicle prior to use of the kit for vaccination, is than added to the lyophilized components in a predetermined amount as written e.g. in the provided technical instructions.
  • inventive mRNA sequence the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine, according to the above described aspects of the invention is provided that can afterwards be used in a method as described above, also.
  • the present invention furthermore provides several applications and uses of the inventive mRNA sequence as defined herei n, of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, of the inventive pharmaceutical composition, of the inventive vaccine, all comprising the inventive mRNA sequence as defined herein or of kits comprising same.
  • the invention provides an mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, and a composition, a pharmaceutical composition, a vaccine and a kit, all comprising the mRNA sequence for use in a method of prophylactic (pre-exposure prophylaxis or post-exposure prophylaxis) and/or therapeutic treatment of Rabies virus infections (rabies). Consequently, in a further aspect, the present invention is directed to the first medical use of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition, the inventive vaccine, and the inventive kit as defined herein as a medicament.
  • the invention provides the use of an mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof as defined above for the preparation of a medicament.
  • the present invention is directed to the second medical use of the mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, as defined herein, optionally in form of a composition comprising a plurality of inventive mRNA sequences, a pharmaceutical composition or vaccine, kit or kit of parts, for the treatment of Rabies virus infections (rabies) as defined herein.
  • the mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof to be used i n a method as said above is a mRNA sequence formulated together with a pharmaceutical ly acceptable vehicle and an optionally additional adjuvant and an optionally additional further component as defined above e.g. a further antigen or a rabies immune globuline.
  • the mRNA sequence used for post-exposure treatment of Rabies virus infection according to the invention can be combined with administration of rabies immune globuline.
  • the inventive mRNA sequence may alternatively be provided such that it is administered for preventing or treating rabies by several doses, each dose containing the inventive mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, e.g. the first dose containing at least one mRNA encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G) (or fragments, variants or derivatives thereof) and the second dose containing at least one mRNA sequence encoding at least one antigenic peptide or protein derived from a different antigen of Rabies virus, preferably from the nucleoprotein N (RAV-N) (or fragments, variants or derivatives thereof).
  • RAV-G glycoprotein G
  • RAV-N nucleoprotein N
  • both doses are administered in a staggered way, i.e. subsequently, shortly one after the other, e.g. within less than 10 minutes, preferably less than 2 minutes, and at the same site of the body to achieve the same immunological effect as for administration of one single composition containing both, e.g. the mRNA encoding the glycoprotein G (RAV-G) and the mRNA encoding the nucleoprotein N (RAV-N).
  • both e.g. the mRNA encoding the glycoprotein G (RAV-G) and the mRNA encoding the nucleoprotein N (RAV-N).
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine may be administered to the patient as a single dose or as at least one single dose, respectively.
  • the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine may be administered to a patient as a single dose followed by a second dose later and optionally even a third, fourth (or more) dose subsequent thereto etc.
  • booster inoculations with the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine may be administered to a patient at specific time intervals, preferably as defined below, following the second (or third, fourth, etc.) inoculation.
  • At least one dose of the inventive mRNA sequence, pharmaceutical composition or vaccine is administered, preferably from 1 to 1 0 doses, more preferably from 2 to 7 doses, even more preferably from 2 to 5 doses and most preferably from 3 to 5 doses.
  • 3 doses are administered.
  • 5 doses are administered.
  • several doses comprise the same mRNA sequence encoding the same antigenic peptide or protein of Rabies virus, e.g. glycoprotein G (RAV-G).
  • the doses are given in a specific time period, e.g. 20-30 or 20-60 days.
  • the interval between the administration of two or more doses is preferably from 5 to 120 days, more preferably from 7 to 15 days or 1 5 to 30 days. In a preferred embodiment, the interval between the administration of two or more doses is at least 7days, more preferably 28 days.
  • prophylaxis at least 5 doses of the inventive mRNA sequence or inventive pharmaceutical composition or vaccine can be administered within 20-30 days.
  • for prophylactic treatment without exposure to the Rabies virus at least 3 doses of the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine can be administered in 20-60 days.
  • a single dose of the inventive mRNA sequence, composition or vaccine comprises a specific amount of the mRNA according to the invention.
  • the inventive mRNA sequence is provided in an amount of at least 40 pg per dose, preferably in an amount of from 40 to 700 pg per dose, more preferably in an amount of from 80 to 400 pg per dose.
  • the amount of the inventive mRNA sequence comprised in a single dose is typically at least 200 pg, preferably from 200 pg to 1 .000 pg, more preferably from 300 pg to 850 pg, even more preferably from 300 pg to 700 pg.
  • the amount of the inventive mRNA sequence comprised in a single dose is typically at least 80 pg, preferably from 80 pg to 700 pg, more preferably from 80 pg to 400 pg.
  • the amount of the inventive mRNA sequence comprised in a single dose is typically at least 80 pg, preferably from 80 pg to 1 .000 pg, more preferably from 80 pg to 850 pg, even more preferably from 80 pg to 700
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal I y, in three doses (40 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (80 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly by jet injection, in three doses (40 g/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the i nventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermally by jet injection, in three doses (80 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermally by jet injection, in three doses (1 60 g dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly by jet injection, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (40 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly in three doses (80 pg/dose), preferably within 20-60 days, e.g.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (640 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (40 g/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on " day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (80 ⁇ g/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (640 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
  • such booster inoculations with the inventive mRNA sequence or inventive pharmaceutical composition or vaccine as disclosed above (second, third etc. vaccination) may utilize an additional compound or component as defined for the inventive mRNA sequence or inventive pharmaceutical composition or vaccine as defined herein.
  • the present invention also provides a method for expression of an encoded antigenic peptide or protein derived from glycoprotein G (RAV-G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus comprising the steps, e.g. a) providing the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, b) applying or administering the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein to an expression system, e.g.
  • inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein
  • it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a pharmaceutical composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the method may be carried out in vitro, in vivo or ex vivo.
  • the method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, preferably Rabies as defined herein.
  • the present invention also provides the use of the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, preferably for diagnostic or therapeutic purposes, for expression of an encoded antigenic peptide or protein, e.g. by applying or administering the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • the use may be applied for laboratory, for research, for diagnostic for commercial production of peptides or proteins and/or for therapeutic purposes.
  • inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein
  • a cell-free expression system typically a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a pharmaceutical composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the use may be carried out in vitro, in vivo or ex vivo.
  • the use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of Rabies virus infections.
  • the invention provides a method of treatment or prophylaxis of rabies virus infections comprising the steps:
  • inventive mRNA sequence the composition comprising a plurality of inventive mRNA sequences, the pharmaceutical composition or the kit or kit of parts comprising the inventive mRNA sequence as defined above;
  • the invention provides in a certain aspect an mRNA sequence comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus.
  • the inventive mRNA sequence is for use in a method of prophylactic and/or therapeutic treatment of infections caused by Rabies viruses.
  • the invention relates to an mRNA sequence as defined herein for use in a method of prophylactic and/or therapeutic treatment of rabies infections.
  • Figure 1 shows the mRNA sequence R2403 according to SEQ ID NO. 24, comprising a
  • FIG. 25 shows the mRNA sequence R2507 according to SEQ ID NO. 25, comprising a
  • 5'-UTR element comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6, a G/C optimized coding region coding for Rabies virus glycoprotein G (RAV-G), the 3'-UTR element albumin7 according to SEQ ID No. 1 8, a poly (A) sequence consisting of 64 adenosines, a poly(C) sequence consisting of 30 cytosines and a histone stem-loop sequence according to SEQ ID No. 27, as comprised in the RAV-G mRNA vaccine.
  • RAV-G Rabies virus glycoprotein G
  • Figure 3 shows that transfection of HeLa cells with mRNA R2403 coding for the RAV-G protein leads to the expression of the encoded RAV-G protein on the cell surface and that the protein is recognized by an anti-RAV-G antibody.
  • R2429 encoding the influenza HA protein of A/Netherlands/602/2009 served as a negative control, as well as untransfected cells. 24 hours post transfection the RAV-G proteins was stained with a rabies specific antibody and FITC-labelled secondary antibody and analysed by FACS as shown in Example 2.
  • Figure 4 shows that RAV-G mRNA vaccine is immunogenic in mice and induces high titers of neutralizing antibodies comparable to licensed vaccines.
  • mice Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine (80 ⁇ g of R2403) or Ringer-Lactate (RiLa) as buffer control. Two groups were intramuscularly (i.m.) injected with 1 /1 0 of the human dose of the licensed vaccines Rabipur® and HDC, respectively. All animals received boost injections on day 21 and blood samples were collected on day 35 for the determination of Rabies virus neutralization titers as described in Example 3.
  • B The RAV-G mRNA vaccine induced long-lasting virus neutralization titers in mice well above the 0.5 lU/ml.
  • Figure 5 shows that RAV-G mRNA vaccine induces antigen-specific CD8 + T cel ls.
  • Example 4 The experiment was performed as describe in Example 4 and T cel ls were analysed by intracellular cytokine staining for the antigen-specific induction of cytokines.
  • Figure 6 shows that RAV-G mRNA vaccine induces antigen-specific CD4 + T cells at significantly higher frequencies than Rabipur®.
  • Example 4 T cells were analysed by intracellular cytokine staining.
  • Figure 7 shows that RAV-G mRNA vaccine induces a dose-dependent functional antibody (virus neutralisation antibody) response in C57BL/6 mice, demonstrating dose-response relationship between RAV-G mRNA and the induction of functional antibodies.
  • Example 5 The experiment was performed as described in Example 5 and virus neutralization titers (lU/ml) were determined.
  • Statistical analysis ANOVA (Kruskal- Wallis), **: p 0.01 ; *: p 0.05.
  • Figure 8 shows that the RAV-G mRNA vaccine protects mice against a lethal rabies virus chal lenge infection.
  • Figure 9 shows that the RAV-G mRNA vaccine protects mice against a lethal virus challenge infection - Influence of immunization schedule
  • Figure 10 shows that the RAV-G mRNA vaccine is stable and immunogenic after storage for 6 months at 40°C or 1 month at 60°C.
  • Figure 1 1 shows that the RAV-G mRNA vaccine induces a protective immune response in adult pigs.
  • Pigs vaccinated with RAV-G mRNA showed a functional antibody response well above the 0.5 lU/ml WHO standard.
  • Figure 12 shows that the RAV-G mRNA vaccine induces significant virus neutralization titers in newborn pigs comparable to a benchmark vaccine (Rabipur®).
  • Figure 13 shows that the RAV-G mRNA vaccine induces virus neutralization titers in mice after intramuscular injection. The experiment was performed as described in
  • Figure 14 shows the induction of virus neutralization titers in mice after intradermal vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 1 1 .
  • Figure 15 shows the induction of virus neutralization titers in domestic pigs after intradermal vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 12.
  • Figure 16 shows the induction of virus neutralization titers in domestic pigs after intramuscular vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 13.
  • DNA sequences, encoding glycoprotein G (RAV-G) of the Pasteur vaccine strain were prepared and used for subsequent in vitro transcription.
  • the corresponding mRNA sequences RAV-G(GC)-muag-A64-C30-histoneSL (R2403) and 32L-RAV-G(GC)-albumin7-A64-C30-histoneSL (R2507) are shown in Figure 1 and 2 according to SEQ. ID No. 24 and 25.
  • the mRNA sequence RAV-G(GC)-muag-A64-C30-histoneSL (R2403; SEQ ID NO:24) comprises in 5'- to 3'-direction:
  • a. a 5'-CAP structure consisting of m7GpppN; b. ) a G/C maximized coding region encoding the full-length protein of RAV-G of the
  • Pasteur vaccine strain according to SEQ ID No. 1 ; c. ) a 3'-UTR element derived from a alpha globin gene, comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22; d. ) a poly(A) sequence, comprising 64 adenosines; e. ) a poly(C) sequence, comprising 30 cytosines; and f.) a histone-stem-loop structure, comprising the RNA sequence according to SEQ ID No 27.
  • R2403 refers to the mRNA sequence, which is defined by the sequence according to SEQ ID NO:24.
  • the "R2403" mRNA may be provided in lyophilised form, which is preferably used for storage and/or transport of the inventive mRNA sequence or may be provided in the solved form in the appropriate liquid.
  • the mRNA according to SEQ ID NO:24 if provided in lyophilised form, is typically reconstituted in an appropriate liquid as defined herein, preferably in Ringer-Lactate, in order to obtain a liquid formulation.
  • the mRNA sequence 32L-RAV-G(GC)-albumin7-A64-C30-histoneSL (R2507) comprises in 5'- to 3'-direction:
  • Pasteur vaccine strain according to SEQ ID No. 1 ; d. ) a 3'UTR element comprising the corresponding RNA sequence of a nucleic acid sequence according to SEQ ID NO. 18; e. ) a poly(A) sequence, comprising 64 adenosines; f. ) a poly(C) sequence, comprising 30 cytosines; and g. ) a histone-stem-loop structure, comprising the RNA sequence according to SEQ
  • the mRNA R2403 or R2507 were complexed with protamine by addition of protamine to the mRNA in the ratio (1 :2) (w/w) (adjuvant component). After incubation for 10 min, the same amount of free mRNA R2403 or R2507 used as antigen-providing mRNA was added.
  • Example 2 In vitro characterization of mRNA encoding Rabies virus G protein (RAV-G)
  • HeLa cells were seeded in a 6-well plate at a density of 300 000 cells/well in cell culture medium (RPMI, 10% FCS, 1 % L-Glutamine, 1 % Pen/Strep) 24h prior to transfection.
  • HeLa cells were transfected with 5 ⁇ g of RAV-G encoding mRNA (R2403) or influenza HA protein of A/Netherlands/602/2009 encoding mRNA (R2429) as negative control using Lipofectamine 2000 (Invitrogen) and stained 24 hours post transfection with a rabies virus specific antibody (HyTest Ltd; #1 1/06-R7-C5) and FITC labelled goat anti-mouse IgG antibody (Invitrogen, #871942A) and analysed by flow cytometry (FACS). The flow cytometry data are evaluated quantitatively by Flowjo software.
  • Figure 3 demonstrates that the RAV-G protein is, as expected for the Rabies G protein, expressed on the surface of transfected cells and can
  • Example 3 Induction of a humoral immune response by the RAV-G mRNA vaccine Immunization
  • mice were intradermally (i.d.) injected with the mRNA vaccine comprising mRNA coding for Rabies virus glycoprotein G (RAV-G) (R2403 according to Example 1 ; 80 ⁇ g/mouse/vacci nation day) or Ringer-lactate (RiLa) as buffer control.
  • RAV-G Rabies virus glycoprotein G
  • RV-G Rabies virus glycoprotein G
  • RV-G Ringer-lactate
  • Two control groups were intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccines Rabipur® (Novartis) and HDC (human diploid cell vaccine, Sanofi Pasteur MSD GmbH), respectively. All animals received boost injections on day 21 and blood samples were collected on day 35 for the determination of virus neutralization titers.
  • the licensed rabies vaccines Rabipur® (Novartis) and HDC (Human diploid cells, Sanofi Pasteur MSD GmbH) comprise inactivated Rabies virus.
  • virus neutralization titer According to WHO standards, an antibody titer is considered protective if the respective VNT is at least 0.5 l U/ml. Therefore, blood samples were taken from vaccinated mice on day 35 and from vaccinated humans on day 42 or as i ndicated after vaccination and sera were prepared. These sera were used in fluorescent antibody virus neutralisation (FAVN) test using the cell culture adapted challenge virus strain (CVS) of rabies virus as recommended by the OIE (World Organisation for Animal Health) and first described in Cliquet F., Aubert M.
  • FAVN fluorescent antibody virus neutralisation
  • Infection of cells is analysed after fixation of cel ls using 80% acetone at room temperature using FITC anti-rabies conjugate. Plates were washed twice using PBS and excess of PBS was removed. Cell cultures are scored positive or negative for the presence of rabies virus. Negative scored cells in sera treated wells represent neutralization of rabies virus.
  • Each FAVN tests includes WHO or OIE standard serum (positive reference serum) that serves as reference for standardisation of the assay. Neutralization activity of test sera is calculated with reference to the standard serum provided by the WHO and displayed as International Units/ml (lU/ml).
  • the RAV-G mRNA vaccine (R2403) induces neutralizing antibody titers comparable to the HDC and Rabipur® vaccines, well above the WHO standard of 0.5 lU/ml.
  • the RAV-G mRNA vaccine induces long-lasting rabies virus neutralization titers in mice.
  • Example 4 Induction of a cellular immune response by the RAV-G mRNA vaccine Immunization
  • mice On day zero, BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 (80 ⁇ g/mouse/vaccination/day) or Ringer-lactate (RiLa) as buffer control.
  • RAV-G mRNA vaccine R2403 80 ⁇ g/mouse/vaccination/day
  • Ringer-lactate Ringer-lactate
  • Intracellular cytokine staining Splenocytes from vaccinated and control mice were isolated according to a standard protocol. Briefly, isolated spleens were grinded through a cell strainer and washed in PBS/1 %FBS followed by red blood cel l lysis. After an extensive washing step with PBS/1 %FBS splenocytes were seeded into 96-well plates (2x1 0 6 cells/well) and kept overnight at 4°C. The next day cells were stimulated with the RAV-G peptide library (J PT) that comprised the amino acid sequence of the Rabies G protein from Pasteur vaccine strain of Rabies virus according to SEQ ID No.
  • J PT RAV-G peptide library
  • the fol lowing antibodies were used for staining: CD8-PECy7 (1 :200), CD3-FITC (1 :200), IL2-PerCP-Cy5.5 (1 :100), TNFa-PE (1 : 100), IFNv-APC (1 :1 00) (eBioscience), CD4-BD Horizon V450 (1 :200) (BD Biosciences) and incubated with FcyR- block diluted 1 :1 00.
  • Aqua Dye was used to distinguish live/dead cells (Invitrogen). Cells were collected using a Canto II flow cytometer (Beckton Dickinson). Flow cytometry data were analysed using Flowjo software (Tree Star, Inc.). Statistical analysis was performed using GraphPad Prism software, Version 5.01 . Statistical differences between groups were assessed by the Mann Whitney test.
  • the RAV-G mRNA vaccine (R2403) induced IFNy positive, TNFa positive and IFNy TNFa double-positive multifunctional CD4 + T cells at significantly higher frequencies than the Rabipur® vaccine which comprises the whole inactivated Rabies virus.
  • Example 5 Induction of a dose-dependent humoral immune response by the RAV-G mRNA vaccine in C57BL/6 mice
  • C57BL/6 mice were intradermally (i.d.) injected with different doses of the RAV-G mRNA vaccine R2403 or Ringer-lactate (RiLa) as buffer control as shown in Table 3.
  • Two groups were intramuscularly (i.m.) injected with 1 /1 0 of the human dose of the licensed vaccines Rabipur® and HDC, respectively. All animals received boost injections on day 21 .
  • Blood samples were taken on day 35 and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3.
  • FAVN fluorescent antibody virus neutralisation
  • mice volume dose schedule
  • the RAV-G mRNA vaccine induces a dose-dependent antibody response.
  • Doses of 20 ⁇ g, 40 ⁇ g and 80 ⁇ g induced significantly higher virus neutralizing antibody titers compared to 0.1 human dose of the HDC vaccine.
  • mice Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 according to the schedule shown in Table 3 or Ringer-lactate (RiLa) as buffer control.
  • Two control groups were intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccines Rabipur® and HDC, respectively.
  • Sixteen days after the last immunization the animals were infected using a 40-fold LD50 dose of the CVS strain of Rabies virus intracranial ly (i.e.). Mice were monitored for specific symptoms of Rabies disease and body weight development.
  • the RAV-G mRNA vaccine protected all mice against a lethal rabies virus challenge infection. All mice vaccinated with RAV-G mRNA or Rabipur® were protected against a lethal challenge infection without weight loss.
  • mice were vaccinated with these samples and their immunogenic and protective potential was evaluated.
  • mice Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 according to the schedule shown in Table 5 or Ringer-lactate (RiLa) as buffer control.
  • a control group was intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccine HDC (stored as recommended by the manufacture at 2-8°C).
  • FAVN fluorescent antibody virus neutralisation
  • the RAV-G mRNA vaccine is stable and immunogenic after storage for 6 months up to 40°C or 1 month at 60°C.
  • the vaccine is still fully protective as demonstrated in a lethal challenge infection (10B)
  • Example 8 Induction of a humoral immune response by the RAV-G mRNA vaccine in pigs Immunization
  • the RAV-G mRNA vaccine induces an immune response after prime / boost vaccination well above the 0.5 lU/ml WHO standard.
  • Example 9 Induction of virus neutralization titers by the RAV-G mRNA vaccine in newborn pigs is comparable to a benchmark vaccine (Rabipur®)
  • the RAV-G mRNA vaccine induces virus neutralizing antibodies after prime-boost vaccination of new born pigs, well above the 0.5 lU/ml WHO standard.
  • Example 10 Induction of virus neutralization titers by the RAV-G mRNA vaccine after intramuscular immunization in mice
  • mice Female BALB/c mice were intramuscularly (i.m.; M. tibialis) injected with the RAV-G mRNA vaccine R2507 or Ringer- lactate (RiLa) as buffer control according to the schedule shown in Table 8.
  • RAV-G mRNA vaccine R2507 or Ringer- lactate (RiLa) as buffer control according to the schedule shown in Table 8.
  • FAVN fluorescent antibody virus neutralisation
  • the RAV-G mRNA vaccine induces virus neutralization titers in mice after intramuscular injection well above the 0.5 lU/ml WHO standard.
  • Example 1 1 Induction of a humoral immune response by the RAV-G mRNA vaccine in mice
  • mice On day zero, BALB/c mice were intradermally (i.d.) injected with the mRNA vaccine comprising mRNA coding for Rabies virus glycoprotein G (RAV-G) (according to Example 1 ; 80 ⁇ g/mouse/vacci nation day). All animals received boost injections on day 7 and 21 .
  • RAV-G Rabies virus glycoprotein G
  • the RAV-G mRNA vaccine (R2403) induces already after two vaccinations neutralizing antibody titers, well above the WHO standard of 0.5 lU/ml.
  • Example 12 Induction of a humoral immune response by the RAV-G mRNA vaccine in domestic pigs
  • Example 13 Induction, of a humoral immune response by the RAV-G mRNA vaccine in domestic pigs Immunization
  • phase I Preliminary results obtained in an ongoing clinical trial demonstrate safety as well as efficacy of the vaccine according to the invention.
  • human volunteers were intradermally injected via jet injection using a Tropis device on day 0, 7 and 28 with the RAV-G mRNA vaccine R2403.
  • the mRNA was prepared as described in Example 1 herein, i.e. mRNA complexed with protamine in a ratio of 2:1 (w/w) was mixed with an equal amount of free mRNA. On each of the three vaccination days, 80 pg of mRNA were administered.
  • VNT virus neutralizing titers

Abstract

The present invention relates to an mRNA sequence, comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof. Additionally the present invention relates to a composition comprising a plurality of mRNA sequences comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof. Furthermore it also discloses the use of the mRNA sequence or the composition comprising a plurality of mRNA sequences for the preparation of a pharmaceutical composition, especially a vaccine, e.g. for use in the prophylaxis or treatment of Rabies virus infections. The present invention further describes a method of treatment or prophylaxis of rabies using the mRNA sequence.

Description

Rabies vaccine
The present invention relates to an mRNA sequence, comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof. Additionally the present invention relates to a composition comprising a plurality of mRNA sequences comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof.
Furthermore it also discloses the use of the mRNA sequence or the composition comprising a plurality of mRNA sequences for the preparation of a pharmaceutical composition, especially a vaccine, e.g. for use in the prophylaxis, postexposure prophylaxis or treatment of Rabies virus infections. The present invention further describes a method of treatment, postexposure prophylaxis or prophylaxis of rabies using the mRNA sequence.
Rabies is a viral zoonosis, endemic in more than 1 00 countries and territories (WHO epidemiological record 2007a. No. 49/50 (82):425-436. Rabies vaccines: WHO position paper), and poses a threat to more than 3 billion people (Knobel DL et al. Re-evaluating the burden of rabies in Africa and Asia. Bulletin of the World Health Organisation 2005;83:360- 368). The disease is invariably fatal following the onset of clinical symptoms occurring in the absence of postexposure prophylaxis (WHO epidemiological record 2010. No. 32 (85):309- 320. Rabies vaccine: WHO position paper). The WHO assumes 55000 rabies related deaths and the postexposure treatment (PET) of more than 1 0 million people each year (WHO Weekly epidemiological record, No. 49/50, 2007b, 82:425-436).
Currently available rabies vaccines include the most widely used but highly risk-prone nerve tissue vaccines, or the safer but more costly cell culture and embryonated egg vaccines (CCEEVs). Risks associated with nerve tissue vaccines include induction of autoimmune central nervous system disease due to their inherent myelin content; the need for multiple injections; and unreliable efficacy (Plotkin SA. Rabies. Clin Infect Dis 2000;30:4-12). The WHO does not recommend the use of nerve tissue vaccines and strongly encourages the increased supply of modern and high quality vaccines to poor populations (WHO epidemiological record 2007a. No. 49/50 (82):425-436. Rabies vaccines: WHO position paper). Avian embryo vaccines and cell culture vaccines contain inactivated purified virus, free from nerve protein. Although safer and more immunogenic than nerve tissue vaccines, cell culture production methods are time-consuming and resource-intensive and the associated cost burden largely restricts the use to the developed world (Warrell MJ and Warrell DA. Intradermal postexposure rabies vaccine regimens. Clin Infect Dis 2000;31 :844- 845) despite of WHO current recommendations.
Pre-exposure prophylaxis (PrEP) with cell culture vaccine is safe, and is recommended for individuals at increased risk (e.g., laboratory staff, veterinarians, animal handlers, wildlife workers and travellers to rabies-endemic areas), but it is largely restricted for reasons of cost to the developed world. Furthermore, the anti-rabies vaccine is recommended for people travelling to countries in Africa and Asia, where rabies is endemic (STI O 201 1 ).
In Germany e.g. only two anti-rabies vaccines are on the market, Rabipur® and "Tollwut- Impfstoff (human diploid cell [HDC]) inaktiviert". These vaccines contain inactivated rabies virus. Both vaccines are recommended for pre- and postexposure use. After contact with rabid or suspected rabid animals, PET (post-exposure treatment) is recommended with vaccinations on days 0, 3, 7, 14 and 28 after exposure. Combined with the vaccination, proper wound management and simultaneous administration of rabies immunoglobulin (Ig) should be performed to effectively prevent the disease.
A current problem is a shortage of these vaccines, which are, at certain times, only available for postexposure prophylaxis and not for prophylactic vaccination. Prophylactic vaccination is, however, important for travellers visiting developing countries where rabies virus Ig for postexposure prophylaxis may not be available.
Therefore there is a need for a save and effective rabies vaccine which can be delivered at any time. Furthermore there is an urgent need for a temperature stabile rabies vaccine which is not dependent on cooling (cold chain).
Furthermore, there is an unmet medical need to improve the effectiveness of rabies vaccine delivery and for the development of a safe and effective rabies vaccine that is more affordable and more rapidly manufactured than the currently available cell culture vaccines.
Therefore it is the object of the underlying invention to provide an mRNA sequence coding for antigenic peptides or proteins of Rabies virus for the use as vaccine for prophylaxis or treatment of rabies, particularly for preexposure prophylaxis or postexposure prophylaxis. Furthermore it is the object of the present invention to provide an effective rabies vaccine which can be stored without cold chain and which enables rapid and scalable vaccine production.
These objects are solved by the subject matter of the attached claims. Particularly, the objects underlying the present invention are solved according to a first aspect by an inventive mRNA sequence comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof.
For the sake of clarity and readability the following scientific background information and definitions are provided. Any technical features disclosed thereby can be part of each and every embodiment of the invention. Additional definitions and explanations can be provided in the context of this disclosure.
Immune system: The immune system may protect organisms from infection. If a pathogen breaks through a physical barrier of an organism and enters this organism, the innate immune system provides an immediate, but non-specific response. If pathogens evade this innate response, vertebrates possess a second layer of protection, the adaptive immune system. Here, the immune system adapts its response during an infection to improve its recognition of the pathogen. This improved response is then retained after the pathogen has been eliminated, in the form of an immunological memory, and allows the adaptive immune system to mount faster and stronger attacks each time this pathogen is encountered. According to this, the immune system comprises the innate and the adaptive immune system. Each of these two parts contains so called humoral and cellular components.
Immune response: An immune response may typically either be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response). The invention relates to the core to specific reactions (adaptive immune responses) of the adaptive immune system. Particularly, it relates to adaptive immune responses to infections by viruses like e.g. Rabies. However, this specific response can be supported by an additional unspecific reaction (innate immune response). Therefore, the invention also relates to a compound for simultaneous stimulation of the innate and the adaptive immune system to evoke an efficient adaptive immune response. Adaptive immune system: The adaptive immune system is composed of highly specialized, systemic cells and processes that eliminate or prevent pathogenic growth. The adaptive immune response provides the vertebrate immune system with the ability to recognize and remember specific pathogens (to generate immunity), and to mount stronger attacks each time the pathogen is encountered. The system is highly adaptable because of somatic hypermutation (a process of increased frequency of somatic mutations), and V(D)J recombination (an irreversible genetic recombination of antigen receptor gene segments). This mechanism allows a small number of genes to generate a vast number of different antigen receptors, which are then uniquely expressed on each individual lymphocyte. Because the gene rearrangement leads to an irreversible change in the DNA of each cell, all of the progeny (offspring) of that cell will then inherit genes encoding the same receptor specificity, including the Memory B cells and Memory T cells that are the keys to long-lived specific immunity. Immune network theory is a theory of how the adaptive immune system works, that is based on interactions between the variable regions of the receptors of T cells, B cells and of molecules made by T cells and B cells that have variable regions.
Adaptive immune response: The adaptive immune response is typically understood to be antigen-specific. Antigen specificity allows for the generation of responses that are tailored to specific antigens, pathogens or pathogen-infected cells. The ability to mount these tailored responses is maintained in the body by "memory cells". Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it. In this context, the first step of an adaptive immune response is the activation of naive antigen- specific T cells or different immune cells able to induce an antigen-specific immune response by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing. Cell types that can serve as antigen-presenting cells are inter alia dendritic cells, macrophages, and B cells. Each of these cells has a distinct function in eliciting immune responses. Dendritic cells take up antigens by phagocytosis and macropinocytosis and are stimulated by contact with e.g. a foreign antigen to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells. Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents or other appropriate stimuli to express MHC molecules. The unique ability of B cells to bind and internalize soluble protein antigens via their receptors may also be important to induce T cells. Presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells. The most important function of effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by Th1 cells which together make up cell-mediated immunity, and the activation of B cells by both Th2 and Th1 cells to produce different classes of antibody, thus driving the humoral immune response. T cells recognize an antigen by their T cell receptors which do not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogen-derived protein antigens, which are bound to MHC molecules on the surfaces of other cells.
Cellular immunity/cellular immune response: Cellular immunity relates typically to the activation of macrophages, natural killer cells (NK), antigen-specific cytotoxic T- lymphocytes, and the release of various cytokines in response to an antigen. In a more general way, cellular immunity is not related to antibodies but to the activation of cells of the immune system. A cellular immune response is characterized e.g. by activating antigen- specific cytotoxic T-lymphocytes that are able to induce apoptosis in body cells displaying epitopes of an antigen on their surface, such as virus-infected cells, cells with intracellular bacteria, and cancer cells displaying tumor antigens; activating macrophages and natural killer cells, enabling them to destroy pathogens; and stimulating cells to secrete a variety of cytokines that influence the function of other cells involved in adaptive immune responses and innate immune responses. Humoral immunity/humoral immune response: Humoral immunity refers typically to antibody production and the accessory processes that may accompany it. A humoral immune response may be typically characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation. Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
Innate immune system: The innate immune system, also known as non-specific immune system, comprises the cells and mechanisms that defend the host from infection by other organisms in a non-specific manner. This means that the cells of the innate system recognize and respond to pathogens in a generic way, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host. The innate immune system may be e.g. activated by ligands of pathogen-associated molecular patterns (PAMP) receptors, e.g. Toll-like receptors (TLRs) or other auxiliary substances such as lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-1 0, IL-12, IL- 13, IL-14, IL-1 5, IL-16, IL-1 7, IL-1 8, IL-1 9, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL- 27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G- CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and hGH, a ligand of human Toll-like receptor TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, a ligand of murine Toll-like receptor TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR13, a ligand of a NOD-like receptor, a ligand of a RIG-I like receptor, an immunostimulatory nucleic acid, an immunostimulatory RNA (isRNA), a CpG-DNA, an antibacterial agent, or an anti-viral agent. Typically a response of the innate immune system includes recruiting immune cells to sites of infection, through the production of chemical factors, including specialized chemical mediators, called cytokines; activation of the complement cascade; identification and removal of foreign substances present in organs, tissues, the blood and lymph, by specialized white blood cells; activation of the adaptive immune system through a process known as antigen presentation; and/or acting as a physical and chemical barrier to infectious agents.
Adjuvant / adjuvant component: An adjuvant or an adjuvant component in the broadest sense is typically a (e.g. pharmacological or immunological) agent or composition that may modify, e.g. enhance, the efficacy of other agents, such as a drug or vaccine. Conventionally the term refers in the context of the invention to a compound or composition that serves as a carrier or auxiliary substance for immunogens and/or other pharmaceutically active compounds. It is to be interpreted in a broad sense and refers to a broad spectrum of substances that are able to increase the immunogenicity of antigens incorporated into or coadministered with an adjuvant in question. In the context of the present invention an adjuvant will preferably enhance the specific immunogenic effect of the active agents of the present invention. Typically, "adjuvant" or "adjuvant component" has the same meaning and can be used mutually. Adjuvants may be divided, e.g., into immuno potentiators, antigenic delivery systems or even combinations thereof.
The term "adjuvant" is typically understood not to comprise agents which confer immunity by themselves. An adjuvant assists the immune system unspecifically to enhance the antigen-specific immune response by e.g. promoting presentation of an antigen to the immune system or induction of an unspecific innate immune response. Furthermore, an adjuvant may preferably e.g. modulate the antigen-specific immune response by e.g. shifting the dominating Th2-based antigen specific response to a more Th1 -based antigen specific response or vice versa. Accordingly, an adjuvant may favourably modulate cytokine expression/secretion, antigen presentation, type of immune response etc.
Immunostimulatory RNA: An immunostimulatory RNA (isRNA) in the context of the invention may typical ly be a RNA that is able to induce an innate immune response itself. It usually does not have an open reading frame and thus does not provide a peptide-antigen or immunogen but elicits an innate immune response e.g. by binding to a specific kind of Tolllike-receptor (TLR) or other suitable receptors. However, of course also mRNAs having an open reading frame and coding for a peptide/protein (e.g. an antigenic function) may induce an i nnate immune response.
Antigen: According to the present invention, the term "antigen" refers typically to a substance which may be recognized by the immune system and may be capable of triggering an antigen-specific immune response, e.g. by formation of antibodies or antigen- specific T-cells as part of an adaptive immune response. An antigen may be a protein or peptide. In this context, the first step of an adaptive immune response is the activation of naive antigen-specific T cells by antigen-presenting cel ls. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing. The three cel l types that can serve as antigen-presenting cel ls are dendritic cells, macrophages, and B cel ls. Each of these cells has a distinct function in eliciting immune responses. Tissue dendritic cel ls take up antigens by phagocytosis and macropinocytosis and are stimulated by infection to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells. Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents to express MHC class II molecules. The unique ability of B cells to bind and internalize soluble protein antigens via their receptors may be important to induce T cells. By presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells. The most important function of effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by TH1 cells which together make up cell-mediated immunity, and the activation of B cells by both TH2 and TH 1 cells to produce different classes of antibody, thus driving the humoral immune response. T cells recognize an antigen by their T cel l receptors which does not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogens' protein antigens, which are bound to MHC molecules on the surfaces of other cel ls. T cells fall into two major classes that have different effector functions. The two classes are distinguished by the expression of the cell-surface proteins CD4 and CD8. These two types of T cells differ in the class of MHC molecule that they recognize. There are two classes of MHC molecules - MHC class I and MHC class II molecules - which differ in their structure and expression pattern on tissues of the body. CD4+ T cells bind to a MHC class II molecule and CD8+ T cells to a MHC class I molecule. MHC class I and MHC class II molecules have distinct distributions among cells that reflect the different effector functions of the T cells that recognize them. MHC class I molecules present peptides of cytosolic and nuclear origin e.g. from pathogens, commonly viruses, to CD8+ T cells, which differentiate into cytotoxic T cells that are specialized to kill any cell that they specifically recognize. Almost all cells express MHC class I molecules, although the level of constitutive expression varies from one cell type to the next. But not only pathogenic peptides from viruses are presented by MHC class I molecules, also self-antigens like tumour antigens are presented by them. MHC class I molecules bind peptides from proteins degraded in the cytosol and transported in the endoplasmic reticulum. The CD8+ T cells that recognize MHC class l:peptide complexes at the surface of infected cells are specialized to kill any cells displaying foreign peptides and so rid the body of cells infected with viruses and other cytosolic pathogens. The main function of CD4+ T cells (CD4+ helper T cells) that recognize MHC class II molecules is to activate other effector cells of the immune system. Thus MHC class II molecules are normally found on B lymphocytes, dendritic cells, and macrophages, cells that participate in immune responses, but not on other tissue cells. Macrophages, for example, are activated to kill the intravesicular pathogens they harbour, and B cells to secrete immunoglobulins against foreign molecules. MHC class II molecules are prevented from binding to peptides in the endoplasmic reticulum and thus MHC class II molecules bind peptides from proteins which are degraded in endosomes. They can capture peptides from pathogens that have entered the vesicular system of macrophages, or from antigens internalized by immature dendritic cells or the immunoglobulin receptors of B cells. Pathogens that accumulate in large numbers inside macrophage and dendritic cell vesicles tend to stimulate the differentiation of TH1 cells, whereas extracellular antigens tend to stimulate the production of TH2 cells. TH1 cells activate the microbicidal properties of macrophages and induce B cells to make lgG antibodies that are very effective of opsonising extracellular pathogens for ingestion by phagocytic cells, whereas TH2 cells initiate the humoral response by activating naive B cells to secrete IgM, and induce the production of weakly opsonising antibodies such as lgG1 and lgG3 (mouse) and lgG2 and lgG4 (human) as well as IgA and IgE (mouse and human). Epitope (also called "antigen determinant"): T cell epitopes or parts of the proteins in the context of the present invention may comprise fragments preferably having a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 1 3 or more amino acids, e.g. 1 3, 14, 15, 1 6, 1 7, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence. These fragments are typically recognized by T cells in form of a complex consisting of the peptide fragment and an MHC molecule.
B cell epitopes are typically fragments located on the outer surface of (native) protein or peptide antigens as defined herein, preferably having 5 to 1 5 amino acids, more preferably having 5 to 12 amino acids, even more preferably having 6 to 9 amino acids, which may be recognized by antibodies, i.e. in their native form.
Such epitopes of proteins or peptides may furthermore be selected from any of the herein mentioned variants of such proteins or peptides. In this context antigenic determinants can be conformational or discontinuous epitopes which are composed of segments of the proteins or peptides as defined herein that are discontinuous in the amino acid sequence of the proteins or peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain. Vaccine: A vaccine is typically understood to be a prophylactic or therapeutic material providing at least one antigen or antigenic function. The antigen or antigenic function may stimulate the body's adaptive immune system to provide an adaptive immune response.
Antigen-providing mRNA: An antigen-providing mRNA in the context of the invention may typically be an mRNA, having at least one open reading frame that can be translated by a cell or an organism provided with that mRNA. The product of this translation is a peptide or protein that may act as an antigen, preferably as an immunogen. The product may also be a fusion protein composed of more than one immunogen, e.g. a fusion protein that consist of two or more epitopes, peptides or proteins derived from the same or different virus-proteins, wherein the epitopes, peptides or proteins may be linked by linker sequences. Bi-/multicistronic mRNA: mRNA, that typically may have two (bicistronic) or more (multicistronic) open reading frames (ORF). An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein. Translation of such a mRNA yields two (bicistronic) or more (multicistronic) distinct translation products (provided the ORFs are not identical). For expression in eukaryotes such mRNAs may for example comprise an internal ribosomal entry site (IRES) sequence.
5'-CAP- Structure: A 5'-CAP is typically a modified nucleotide, particularly a guanine nucleotide, added to the 5' end of an mRNA-molecule. Preferably, the 5'-CAP is added using a 5 '-5 '-triphosphate linkage (also named m7GpppN). Further examples of 5'-CAP structures include glyceryl, inverted deoxy abasic residue (moiety), 4', 5' methylene nucleotide, l -(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, 1 ,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4- dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3 '-3 '-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3 '-2 '-inverted nucleotide moiety, 3'-2'- inverted abasic moiety, 1 ,4-butanediol phosphate, 3'-phosphoramidate, hexylphosphate, aminohexyl phosphate, 3'-phosphate, 3'phosphorothioate, phosphorodithioate, or bridging or non-bridging methylphosphonate moiety. These modified 5'-CAP structures may be used in the context of the present invention to modify the inventive mRNA sequence. Further modified 5'-CAP structures which may be used in the context of the present invention are CAP1 (methylation of the ribose of the adjacent nucleotide of m7GpppN), CAP2 (methylation of the ribose of the 2nd nucleotide downstream of the m7GpppN), CAP3 (methylation of the ribose of the 3rd nucleotide downstream of the m7GpppN), CAP4 (methylation of the ribose of the 4th nucleotide downstream of the m7GpppN), ARCA (anti- reverse CAP analogue, modified ARCA (e.g. phosphothioate modified ARCA), inosine, N1 - methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino- guanosine, LNA-guanosine, and 2-azido-guanosine.
Fragments of proteins: "Fragments" of proteins or peptides in the context of the present invention may, typically, comprise a sequence of a protein or peptide as defined herein, which is, with regard to its amino acid sequence (or its encoded nucleic acid molecule), N- terminally and/or C-terminally truncated compared to the amino acid sequence of the original (native) protein (or its encoded nucleic acid molecule). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level. A sequence identity with respect to such a fragment as defined herein may therefore preferably refer to the entire protein or peptide as defined herein or to the entire (coding) nucleic acid molecule of such a protein or peptide.
Fragments of proteins or peptides in the context of the present invention may furthermore comprise a sequence of a protein or peptide as defined herein, which has a length of for example at least 5 amino acids, preferably a length of at least 6 amino acids, preferably at least 7 amino acids, more preferably at least 8 amino acids, even more preferably at least 9 amino acids; even more preferably at least 10 amino acids; even more preferably at least 1 1 amino acids; even more preferably at least 12 amino acids; even more preferably at least 13 amino acids; even more preferably at least 14 amino acids; even more preferably at least 1 5 amino acids; even more preferably at least 1 6 amino acids; even more preferably at least 1 7 amino acids; even more preferably at least 1 8 amino acids; even more preferably at least 19 amino acids; even more preferably at least 20 amino acids; even more preferably at least 25 amino acids; even more preferably at least 30 amino acids; even more preferably at least 35 amino acids; even more preferably at least 50 amino acids; or most preferably at least 100 amino acids. For example such fragment may have a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 1 3 or more amino acids, e.g. 13, 14, 1 5, 1 6, 1 7, 1 8, 1 9, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence. These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognized in their native form. Fragments of proteins or peptides may comprise at least one epitope of those proteins or peptides. Furthermore also domains of a protein, like the extracellular domain, the intracellular domain or the transmembrane domain and shortened or truncated versions of a protein may be understood to comprise a fragment of a protein.
Variants of proteins: "Variants" of proteins or peptides as defined in the context of the present invention may be generated, having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s). Preferably, these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property. "Variants" of proteins or peptides as defined in the context of the present invention may comprise conservative amino acid substitution(s) compared to their native, i.e. non-mutated physiological, sequence. Those amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined herein. Substitutions in which amino acids, which originate from the same class, are exchanged for one another are called conservative substitutions. In particular, these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function. This means that e.g. an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g. serine (threonine) by threonine (serine) or leucine (isoleucine) by isoleucine (leucine)). Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1 985, Absorption, Circular Dichroism and ORD of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger et a/, (ed.), Elsevier, Amsterdam).
A "variant" of a protein or peptide may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid identity over a stretch of 10, 20, 30, 50, 75 or 100 amino acids of such protein or peptide.
Furthermore, variants of proteins or peptides as defined herein, which may be encoded by a nucleic acid molecule, may also comprise those sequences, wherein nucleotides of the encoding nucleic acid sequence are exchanged according to the degeneration of the genetic code, without leading to an alteration of the respective amino acid sequence of the protein or peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutation(s) within the above meaning. Identity of a sequence: In order to determine the percentage to which two sequences are identical, e.g. nucleic acid sequences or amino acid sequences as defined herein, preferably the amino acid sequences encoded by a nucleic acid sequence of the polymeric carrier as defined herein or the amino acid sequences themselves, the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. a position of a first sequence may be compared with the corresponding position of the second sequence. If a position in the first sequence is occupied by the same component (residue) as is the case at a position in the second sequence, the two sequences are identical at this position. If this is not the case, the sequences differ at this position. If insertions occur in the second sequence in comparison to the first sequence, gaps can be inserted into the first sequence to allow a further alignment. If deletions occur in the second sequence in comparison to the first sequence, gaps can be inserted into the second sequence to allow a further alignment. The percentage to which two sequences are identical is then a function of the number of identical positions divided by the total number of positions including those positions which are only occupied in one sequence. The percentage to which two sequences are identical can be determined using a mathematical algorithm. A preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. (1 993), PNAS USA, 90:5873-5877 or Altschul et al. (1 997), Nucleic Acids Res., 25:3389-3402. Such an algorithm is integrated in the BLAST program. Sequences which are identical to the sequences of the present invention to a certain extent can be identified by this program.
Derivative of a protein or peptide: A derivative of a peptide or protein is typically understood to be a molecule that is derived from another molecule, such as said peptide or protein. A "derivative" of a peptide or protein also encompasses fusions comprising a peptide or protein used in the present invention. For example, the fusion comprises a label, such as, for example, an epitope, e.g., a FLAG epitope or a V5 epitope. For example, the epitope is a FLAG epitope. Such a tag is useful for, for example, purifying the fusion protein.
Monocistronic mRNA: A monocistronic mRNA may typically be an mRNA, that encodes only one open reading frame. An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein.
Nucleic acid: The term nucleic acid means any DNA- or RNA-molecule and is used synonymous with polynucleotide. Wherever herein reference is made to a nucleic acid or nucleic acid sequence encoding a particular protein and/or peptide, said nucleic acid or nucleic acid sequence, respectively, preferably also comprises regulatory sequences allowing in a suitable host, e.g. a human being, its expression, i.e. transcription and/or translation of the nucleic acid sequence encoding the particular protein or peptide.
Peptide: A peptide is a polymer of amino acid monomers. Usually the monomers are linked by peptide bonds. The term "peptide" does not limit the length of the polymer chain of amino acids. In some embodiments of the present invention a peptide may for example contain less than 50 monomer units. Longer peptides are also called polypeptides, typically having 50 to 600 monomeric units, more specifically 50 to 300 monomeric units. Pharmaceutically effective amount: A pharmaceutically effective amount in the context of the invention is typically understood to be an amount that is sufficient to induce an immune response.
Protein: A protein typically consists of one or more peptides and/or polypeptides folded into 3-dimensional form, facilitating a biological function.
Poly (C) sequence: A poly-(C)-sequence is typically a long sequence of cytosine nucleotides, typically about 10 to about 200 cytosine nucleotides, preferably about 10 to about 100 cytosine nucleotides, more preferably about 10 to about 70 cytosine nucleotides or even more preferably about 20 to about 50 or even about 20 to about 30 cytosine nucleotides. A poly(C) sequence may preferably be located 3' of the coding region comprised by a nucleic acid.
Poly-A-tail: A poly-A-tail also called "3'-poly(A) tail" is typically a long sequence of adenosine nucleotides of up to about 400 adenosine nucleotides, e.g. from about 25 to about 400, preferably from about 50 to about 400, more preferably from about 50 to about 300, even more preferably from about 50 to about 250, most preferably from about 60 to about 250 adenosine nucleotides, added to the 3' end of a RNA. Stabilized nucleic acid: A stabilized nucleic acid, typically, exhibits a modification increasing resistance to in vivo degradation (e.g. degradation by an exo- or endo-nuclease) and/or ex vivo degradation (e.g. by the manufacturing process prior to vaccine administration, e.g. in the course of the preparation of the vaccine solution to be administered). Stabilization of RNA can, e.g., be achieved by providing a 5'-CAP- Structure, a Poly-A-Tail, or any other UTR-modification. It can also be achieved by backbone- modification or modification of the G/C-content of the nucleic acid. Various other methods are known in the art and conceivable in the context of the invention.
Carrier / polymeric carrier: A carrier in the context of the invention may typically be a compound that facilitates transport and/or complexation of another compound. Said carrier may form a complex with said other compound. A polymeric carrier is a carrier that is formed of a polymer.
Cationic component: The term "cationic component" typically refers to a charged molecule, which is positively charged (cation) at a pH value of typically about 1 to 9, preferably of a pH value of or below 9 (e.g. 5 to 9), of or below 8 (e.g. 5 to 8), of or below 7 (e.g. 5 to 7), most preferably at physiological pH values, e.g. about 7.3 to 7.4. Accordingly, a cationic peptide, protein or polymer according to the present invention is positively charged under physiological conditions, particularly under physiological salt conditions of the cell in vivo. A cationic peptide or protein preferably contains a larger number of cationic amino acids, e.g. a larger number of Arg, His, Lys or Orn than other amino acid residues (in particular more cationic amino acids than anionic amino acid residues like Asp or Glu) or contains blocks predominantly formed by cationic amino acid residues. The definition "cationic" may also refer to "polycationic" components.
Vehicle: An agent, e.g. a carrier, that may typically be used within a pharmaceutical composition or vaccine for facilitating administering of the components of the pharmaceutical composition or vaccine to an individual. 3'-untranslated region (3'UTR): A 3'UTR is typically the part of an mRNA which is located between the protein coding region (i.e. the open reading frame) and the poly(A) sequence of the mRNA. A 3'UTR of the mRNA is not translated into an amino acid sequence. The 3'UTR sequence is generally encoded by the gene which is transcribed into the respective mRNA during the gene expression process. The genomic sequence is first transcribed into pre-mature mRNA, which comprises optional introns. The pre-mature mRNA is then further processed into mature mRNA in a maturation process. This maturation process comprises the steps of 5'-Capping, splicing the pre-mature mRNA to excise optional introns and modifications of the 3'-end, such as polyadenylation of the 3'-end of the premature mRNA and optional endo- or exonuclease cleavages etc. In the context of the present invention, a 3'UTR corresponds to the sequence of a mature mRNA which is located 3' to the stop codon of the protein coding region, preferably immediately 3' to the stop codon of the protein coding region, and which extends to the 5'-side of the poly(A) sequence, preferably to the nucleotide immediately 5' to the poly(A) sequence. The term "corresponds to" means that the 3'UTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining the 3'UTR sequence, or a DNA sequence which corresponds to such RNA sequence. In the context of the present invention, the term "a 3'UTR of a gene", such as "a 3'UTR of an albumin gene", is the sequence which corresponds to the 3'UTR of the mature mRNA derived from this gene, i.e. the mRNA obtained by transcription of the gene and maturation of the pre-mature mRNA. The term "3'UTR of a gene" encompasses the DNA sequence and the RNA sequence of the 3'UTR.
5'-untranslated region (5'UTR): A 5'-UTR is typically understood to be a particular section of messenger RNA (mRNA). It is located 5' of the open reading frame of the mRNA. Typically, the 5'UTR starts with the transcriptional start site and ends one nucleotide before the start codon of the open reading frame. The 5'-UTR may comprise elements for controlling gene expression, also called regulatory elements. Such regulatory elements may be, for example, ribosomal binding sites or a 5'-Terminal Oligopyrimidine Tract. The 5'UTR may be posttranscriptionally modified, for example by addition of a 5'-CAP. In the context of the present invention, a 5'UTR corresponds to the sequence of a mature mRNA which is located between the 5'-CAP and the start codon. Preferably, the 5'UTR corresponds to the sequence which extends from a nucleotide located 3' to the 5'-CAP, preferably from the nucleotide located immediately 3' to the 5'-CAP, to a nucleotide located 5' to the start codon of the protein coding region, preferably to the nucleotide located immediately 5' to the start codon of the protein coding region. The nucleotide located immediately 3' to the 5'-CAP of a mature mRNA typically corresponds to the transcriptional start site. The term "corresponds to" means that the 5'UTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining the 5'UTR sequence, or a DNA sequence which corresponds to such RNA sequence. In the context of the present invention, the term "a 5'UTR of a gene", such as "a 5'UTR of a TOP gene", is the sequence which corresponds to the 5'UTR of the mature mRNA derived from this gene, i.e. the mRNA obtained by transcription of the gene and maturation of the pre-mature mRNA. The term "5'UTR of a gene" encompasses the DNA sequence and the RNA sequence of the 5'UTR.
5'Terminal Oligopyrimidine Tract (TOP): The 5'terminal oligopyrimidine tract (TOP) is typically a stretch of pyrimidine nucleotides located at the 5' terminal region of a nucleic acid molecule, such as the 5' terminal region of certain mRNA molecules or the 5' terminal region of a functional entity, e.g. the transcribed region, of certain genes. The sequence starts with a cytidine, which usually corresponds to the transcriptional start site, and is followed by a stretch of usually about 3 to 30 pyrimidine nucleotides. For example, the TOP may comprise 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 1 3, 14, 1 5, 1 6, 1 7, 18, 1 9, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30 or even more nucleotides. The pyrimidine stretch and thus the 5' TOP ends one nucleotide 5' to the first purine nucleotide located downstream of the TOP. Messenger RNA that contains a 5'terminal oligopyrimidine tract is often referred to as TOP mRNA. Accordingly, genes that provide such messenger RNAs are referred to as TOP genes. TOP sequences have, for example, been found in genes and mRNAs encoding peptide elongation factors and ribosomal proteins.
TOP motif: In the context of the present invention, a TOP motif is a nucleic acid sequence which corresponds to a 5'TOP as defined above. Thus, a TOP motif in the context of the present invention is preferably a stretch of pyrimidine nucleotides having a length of 3-30 nucleotides. Preferably, the TOP-motif consists of at least 3 pyrimidine nucleotides, preferably at least 4 pyrimidine nucleotides, preferably at least 5 pyrimidine nucleotides, more preferably at least 6 nucleotides, more preferably at least 7 nucleotides, most preferably at least 8 pyrimidine nucleotides, wherein the stretch of pyrimidine nucleotides preferably starts at its 5'end with a cytosine nucleotide. In TOP genes and TOP mRNAs, the TOP-motif preferably starts at its 5'end with the transcriptional start site and ends one nucleotide 5' to the first purin residue in said gene or mRNA. A TOP motif in the sense of the present invention is preferably located at the 5'end of a sequence which represents a 5'UTR or at the 5'end of a sequence which codes for a 5'UTR. Thus, preferably, a stretch of 3 or more pyrimidine nucleotides is called "TOP motif" in the sense of the present invention if this stretch is located at the 5'end of a respective sequence, such as the inventive mRNA, the 5'UTR element of the inventive mRNA, or the nucleic acid sequence which is derived from the 5'UTR of a TOP gene as described herein. In other words, a stretch of 3 or more pyrimidine nucleotides which is not located at the 5'-end of a 5'UTR or a 5'UTR element but anywhere within a 5'UTR or a 5'UTR element is preferably not referred to as "TOP motif".
TOP gene: TOP genes are typically characterised by the presence of a 5' terminal oligopyrimidine tract. Furthermore, most TOP genes are characterized by a growth- associated translational regulation. However, also TOP genes with a tissue specific translational regulation are known. As defined above, the 5'UTR of a TOP gene corresponds to the sequence of a 5'UTR of a mature mRNA derived from a TOP gene, which preferably extends from the nucleotide located 3' to the 5'-CAP to the nucleotide located 5' to the start codon. A 5'UTR of a TOP gene typically does not comprise any start codons, preferably no upstream AUGs (uAUGs) or upstream open reading frames (uORFs). Therein, upstream AUGs and upstream open reading frames are typically understood to be AUGs and open reading frames that occur 5' of the start codon (AUG) of the open reading frame that should be translated. The 5'UTRs of TOP genes are generally rather short. The lengths of 5'UTRs of TOP genes may vary between 20 nucleotides up to 500 nucleotides, and are typically less than about 200 nucleotides, preferably less than about 1 50 nucleotides, more preferably less than about 100 nucleotides. Exemplary 5'UTRs of TOP genes in the sense of the present invention are the nucleic acid sequences extending from the nucleotide at position 5 to the nucleotide located immediately 5' to the start codon (e.g. the ATG) in the sequences according to SEQ ID Nos. 1 -1363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422of the patent application WO201 3/143700 or homologs or variants thereof, whose disclosure is incorporated herewith by reference. In this context a particularly preferred fragment of a 5'UTR of a TOP gene is a 5'UTR of a TOP gene lacking the 5'TOP motif. The term '5'UTR of a TOP gene' preferably refers to the 5'UTR of a naturally occurring TOP gene.
Fragment of a nucleic acid sequence, particularly an mRNA: A fragment of a nucleic acid sequence consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length nucleic acid sequence which is the basis for the nucleic acid sequence of the fragment, which represents at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length nucleic acid sequence. Such a fragment, in the sense of the present invention, is preferably a functional fragment of the full-length nucleic acid sequence.
Variant of a nucleic acid sequence, particularly an mRNA: A variant of a nucleic acid sequence refers to a variant of nucleic acid sequences which forms the basis of a nucleic acid sequence. For example, a variant nucleic acid sequence may exhibit one or more nucleotide deletions, insertions, additions and/or substitutions compared to the nucleic acid sequence from which the variant is derived. Preferably, a variant of a nucleic acid sequence is at least 40%, preferably at least 50%, more preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, even more preferably at least 90%, most preferably at least 95% identical to the nucleic acid sequence the variant is derived from. Preferably, the variant is a functional variant. A "variant" of a nucleic acid sequence may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% nucleotide identity over a stretch of 1 0, 20, 30, 50, 75 or 1 00 nucleotide of such nucleic acid sequence.
Homolog of a nucleic acid sequence: The term "homolog" of a nucleic acid sequence refers to sequences of other species than the particular sequence. It is particular preferred that the nucleic acid sequence is of human origin and therefore it is preferred that the homolog is a homolog of a human nucleic acid sequence.
Jet injection: The term "jet injection", as used herein, refers to a needle-free injection method, wherein a fluid containing at least one inventive mRNA sequence and, optional ly, further suitable excipients is forced through an orifice, thus generating an ultra-fine liquid stream of high pressure that is capable of penetrating mammalian skin and, depending on the injection settings, subcutaneous tissue or muscle tissue. In principle, the liquid stream forms a hole in the skin, through which the liquid stream is pushed into the target tissue. Preferably, jet injection is used for intradermal, subcutaneous or intramuscular injection of the mRNA sequence according to the invention. In a preferred embodiment, jet injection is used for intramuscular injection of the mRNA sequence according to the invention. In a further preferred embodiment, jet injection is used for intradermal injection of the mRNA sequence according to the invention.
The present invention is based on the surprising finding of the present inventors that an mRNA sequence comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus induces antigen-specific immune responses which neutralize Rabies virus particles and therefore prevent Rabies virus infections. It was very surprising for the inventors that the inventive mRNA sequence induces at least the same immune responses than a licensed rabies vaccine which consists of the whole inactived rabies virus. Even more surprisingly the inventive mRNA sequence coding for an antigenic protein of Rabies virus induced higher antigen-specific CD4+-T cells than a licenced rabies vaccine.
Furthermore, the inventors surprisingly found that the mRNA-based rabies vaccine accordi ng to the invention was biologically active after storage at 40°C for 6 months and even after storage at 60°C for 1 month. Therefore the mRNA-based rabies vaccine according to the invention would be an attractive option for postexposure prophylaxis in developing countries, since it can be stored at ambient temperature, compared to the licenced vaccines which have to be stored between +2 and +8°C. In summary the inventive mRNA sequence comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus could contribute to affordable, readily available, temperature-stable rabies vaccines, "particularly for preexposure and postexposure rabies prophylaxis for the developed and developing world.
Additionally, the mRNA sequence according to the invention enables rapid and rational vaccine design with flexibility, speed and scalability of production probably exceeding those of current virus-based technologies.
In this context it is particularly preferred that the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof. The coding region of the inventive mRNA sequence according to the first aspect of the present invention may occur as a mono-, di-, or even multicistronic mRNA, i.e. an mRNA sequence which carries the coding sequences of one, two or more proteins or peptides. Such coding sequences in di-, or even multicistronic mRNAs may be separated by at least one internal ribosome entry site (IRES) sequence, e.g. as described herein or by signal peptides which induce the cleavage of the resulting polypeptide which comprises several proteins or peptides.
According to the first aspect of the present invention, the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof.
In a particularly preferred embodiment of the first aspect of the invention the inventive mRNA sequence comprises a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G) of Rabies virus or a fragment, variant or derivative thereof.
In this context the amino acid sequence of the at least one antigenic peptide or protein may be selected from any peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), and the RNA polymerase L (RAV-L) of any Rabies virus isolate or a fragment, variant or derivative thereof or from any synthetically engineered rabies peptide or protein.
In a particularly preferred embodiment the full-length protein of the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) is encoded by the coding region comprised in the inventive mRNA.
In a further particularly preferred embodiment a fragment comprising at least one epitope of the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) is encoded by the coding region comprised in the inventive mRNA.
Particularly preferred are the amino acid sequences of a Rabies vaccine strain, preferably of the Pasteur vaccine strain according to the NCBI accession No. M13215: Glycoprotein G (RAV-G) of Pasteur vaccine strain:
Amino acid sequence according to SEQ ID No.1 :
MVPQALLFVPLLVFPLCFGKFPIYTIPD LGPWSPIDIHHLSCPNNLVVEDEGCTNLSGFSYMELK VGYISAIKMNGFTCTGVVTEAETYTNFVGYVTTTFKR HFRPTPDACRAAYNW MAGDPRYEE SLHNPYPDYHWLRTVKTTKESLVIISPSVADLDPYDRSLHSRVFPGGNCSGVAVSSTYCSTNHD YTIWMPENPRLGMSCDIFTNSRG RAS GSETCGFVDERGLYKSLKGACKLKLCGVLGLRLMD GTWVAMQTSNET WCPPGQLVNLHDFRSDEIEHLVVEELVK REECLDALESIMTTKSVSFRRL SHLRKLVPGFGKAYTIFNKTLMEADAHYKSVRTWNEIIPS GCLRVGGRCHPHVNGVFFNGIIL GPDGNVLIPEMQSSLLQQHMELLVSSVIPLMHPLADPSTVF NGDEAEDFVEVHLPDVHERISG VDLGLPNWGKYVLLSAGALTALMLIIFLMTCWRRVNRSEPTQHNLRGTGREVSVTPQSG IISS WESYKSGGETGL
Nucleoprotein N (RAV-N) of Pasteur vaccine strain:
Amino acid sequence according to SEQ ID No.2:
MDAD IVFKVNNQVVSLKPEIIVDQYEYKYPAIKDL KPCITLGKAPDLN AYKSVLSCMSAAKL DPDDVCSYLAAAMQFFEGTCPEDWTSYGIVIARKGDKITPGSLVEIKRTDVEGNWALTGGMEL TRDPTVPEHASLVGLLLSLYRLSKISGQSTGNYKTNIADRIEQIFETAPFV IVEHHTLMTTHKMC ANWSTIPNFRFLAGTYDMFFSRIEHLYSAIRVGTVVTAYEDCSGLVSFTGFIKQINLTAREAILYFF H NFEEEIRRMFEPGQETAVPHSYFIHFRSLGLSG SPYSSNAVGHVFNLIHFVGCYMGQVRSL NATVIAACAPHEMSVLGGYLGEEFFGKGTFERRFFRDE ELQEYEAAELT TDVALADDGTVNS DDEDYFSGETRSPEAVYTRIIMNGGRLKRSHIRRYVSVSSNHQARPNSFAEFLNKTYSSDS Phosphoprotein P (also named M1 ) (RAV-P) of Pasteur vaccine strain:
Amino acid sequence according to SEQ ID No.3:
MSKIFVNPSAIRAGLADLEMAEETVDLINRNIEDNQAHLQGEPIEVDNLPEDMGRLHLDDGKSP NPGEMAKVGEG YREDFQMDEGEDPSLLFQSYLDNVGVQIVRQIRSGERFL IWSQTVEEIISY VAVNFPNPPGKSSED STQTTGRELKKETTPTPSQRESQSSKARMAAQTASGPPALEWSATNEE DDLSVEAEIAHQIAESFS KYKFPSRSSGILLYNFEQLKMNLDDIV EAKNVPGVTRLARDGSKLP LRCVLGWVALANSK FQLLVESNKLSKIMQDDLNRYTSC Matrix protein M (also named M2) (RAV-M) of Pasteur vaccine strain:
Amino acid sequence according to SEP ID No.4:
MNFLRKIVKNCRDEDTQKPSPVSAPLDDDDLWLPPPEYVPLKELTSKKNRRNFCINGGVKVCSP NGYSFGILRHILRSFDEIYSGNHRMVGLV VVIGLALSGAPVPEGMNWVYKLRRTLIFQWADSR GPLEGEELEYSQEITWDDNTEFVGLQIRVSAKQCHIRGRIWCINMNSRAGQLWSDMSLQTQR SEEDKDSSLLLE
RNA Polymerase L (RAV-L) of Pasteur vaccine strain:
Amino acid sequence according to SEQ ID No.5:
MLDPGEVYDDPIDPIELEAEPRGTPTVPNILRNSDYNLNSPLIEDPARLMLEWLKTGNRPYRMTL TDNCSRSFRVLKDYFKKVDLGSL VGGMAAQSMISLWLYGAHSESNRSRRCITDLAHFYSKSSPI E LLNLTLGNRGLRIPPEGVLSCLERVDYDNAFGRYLANTYSSYLFFHVITLYMNALDWDEEKTIL ALW DLTSVDIGKDLVKFKDQIWGLLIVTKDFVYSQSSNCLFDRNYTLML DLFLSRFNSLMVL LSPPEPRYSDDLISQLCQLYIAGDQVLSMCGNSGYEVI ILEPYVVNSLVQRAEKFRPLIHSLGDF PVFIKDKVSQLEETFGSCARRFFRALDQFDNIHDLVFVYGCYRHWGHPYIDYRKGLS LYDQV HIK VIDKSYQECLASDLARRILRWGFD YS WYLDSRFLARDHPLTPYIKTQTWPPKHIVDLV GDTWHKLPITQIFEIPESMDPSEILDDKSHSFTRTRLASWLSENRGGPVPSEKVIITALS PPVNPRE FLKSIDLGGLPDEDLIICL PKEREL IEGRFFALMSWNLRLYFVITE LLANYILPLFDALTMTDNL NKVFKKLIDRVTGQGLLDYSRVTYAFHLDYE WNNHQRLESTEDVFSVLDQVFGLKRVFSRTH EFFQKSWIYYSDRSDLIGLREDQIYCLDASNGPTCWNGQDGGLEGLRQKGWSLVSLLMIDRES QIRNTRT VLAQGDNQVLCPTYMLSPGLSQEGLLYELESISRNAFSIYRAVEEGASKLGLIIKKEET MCSYDFLIYGKTPLFRGNILVPESKRWARVSCVSNDQIVNLANIMSTVSTNALTVAQHSQSLIKP MRDFLLMSVQAVFHYLLFSPIL GRVYKILSAEGESFLLAMSRIIYLDPSLGGVSGMSLGRFHIRQF SDPVSEGLSFWREIWLSSHESWIHALCQEAGNPDLGERTLESFTRLLEDPTTLNIRGGASPTILLK DAIRKALYDEVDKVENSEFREAILLSKTHRDNFILFLTSVEPLFPRFLSELFSSSFLGIPESIIGLIQNSRT IRRQFRKSLS TLEESFYNSEIHGISRMTQTPQRVGGVWPCSSERADLLREISWGRKVVGTTVPH PSEMLGLLPKSSISCTCGATGGGNPRVSVSVLPSFDQSFFCTGPL GYLGSSTSMSTQLFHAWEK VTNVHVV RALSL ESINWFITRDSNLAQTLIRNIVSLTGPDFPLEEAPVF RTGSALHRF SARYS EGGYSSVCPNLLSHISVSTDTMSDLTQDG NYDFMFQPLMLYAQTWTSELVQRDTRLRDSTF HWHLQCNRCVRPIDDVTLETSQIFEFPDVS RISRMVSGAVPHFQRLPDIRLRPGDFESLSGREK SHHIGSAQGLLYSILVAIHDSGYNDGTIFPVNIYGKVSPRDYLRGLARGVLIGSSICFLTRMTNINI NRPLELISGVISYILLRLDNHPSLYIMLREPSFREEIFSIPQ IPAAYPTTMKEGNRSILCYLQHVLRYE REVITASPENDWLWIFSDFRSA MTYLTLITYQSHLLLQRVERNLSKSMRDNLRQLSSLMRQVLG GHGEDTLESDDNIQRLL DSLRRTRWVDQEVRHAARTMTGDYSPNKKVSRKVGCSEWVCSA QQVAVSTSANPAPVSELDIRALS RFQNPLISGLRVVQWATGAHYKL PILDDLNVFPSLCLVV GDGSGGISRAVLNMFPDA LVFNSLLEVNDLMASGTHPLPPSAIMRGGNDIVSRVIDFDSIWEK PSDLRNLATWKYFQSVQKQVNMSYDLIICDAEVTDIASINRITLLMSDFALSIDGPLYLVFKTYGT MLVNPNYKAIQHLSRAFPSVTGFITQVTSSFSSELYLRFS RGKFFRDAEYLTSSTLREMSLVLFNCS SPKSEMQRARSLNYQDLVRGFPEEIISNPYNEMIITLIDSDVESFLVHKMVDDLELQRGTLSKVAIII AIMIVFSNRVFNVSKPLTDPLFYPPSDPKILRHFNICCSTMMYLSTALGDVPSFARLHDLYNRPITY YFRKQVILGNVYLSWSWSNDTSVFKRVACNSSLSLSSHWIRLIYKIVKTTRLVGSI DLSGEVERH LHRYNRWITLENIRSRSSLLDYSCLCIGYSW PAHAKTLV
Additionally particularly preferred are the amino acid sequences of the Flury-LEP vaccine strain (used for Rabipur®) according to the NCBI accession No. GU565703:
Glycoprotein G (RAV-G) of Flury-LEP vaccine strain:
Amino acid sequence according to SEP ID No. 6:
MVPQVLLFVPLLGFSLCFGKFPIYTIPDKLGPWSPIDIHHLSCPNNLVVEDEGCTNLSEFSYMELK VGYISAIKVNGFTCTGVVTEAETYTNFVGYVTTTFKR HFRPTPDACRAAYNWKMAGDPRYEE SLHNPYPDYHWLRTV TT ESLVIISPSVTDLDPYD SLHSRVFPGGNCSGITVSSTYCSTNHDY TIWMPENLRLGTSCDIFTNSRG RASKGGKTCGFVDERGLYKSL GACKLKLCGVLGLRLMDG TWVAMQTSDETKWCPPGQLVNLHDFRSDEIEHLVVEELVKKREECLDALESIMTTKSVSFRRLS HLRKLVPGFG AYT1FNKTLMEADAHYKSVRTWNEIIPS GCLRVGGRCHPHVNGVFFNGIILG SDGHVLIPEMQSSLLQQHMELLESSVIPLMHPLADPSTVFKDGDEVEDFVEVHLPDVHEQVSG VELGLPNWG YVLMIAGALIALMLIIFLMTCCRRVNRPESTQSSLGETGRNVSVTSQSG VISSW ESYKSGGETRL Nucleoprotein N (Rav N) of Flury-LEP vaccine strain:
Amino acid sequence according to SEP ID No. 7:
MDADKIVFKVNNQVVSL PEIIVDQYEY YPAIKDLK PCITLG APDLNKAYKSVLSGMNAA LDPDDVCSYLAAAMQFFEGTCPEDWTSYGILIARKGDKITPDSLVEIKRTDVEGNWALTGGME LTRDPTVSEHASLVGLLLSLYRLSKISGQNTGNY TNIADRIEQIFETAPFVKIVEHHTLMTTH M CANWSTIPNFRFLAGTYDMFFSRIEHLYSAIRVGTVVTAYEDCSGLVSFTGFI QINLTAREAILYF FHKNFEEEIRRMFEPGQETAVPHSYFIHFRSLGLSG SPYSSNAVGHVFNLIHFVGCYMGQVRSL NATVIAACAPHEMSVLGGYLGEEFFG GTFERRFFRDE ELQEYEAAELTKTDVALADDGTVNS DDEDYFSGETRSPEAVYTRIMMNGGRL RSHIRRYVSVSSNHQARPNSFAEFLNKTYSSDS
Phosphoprotein P (Rav P) of Flury-LEP vaccine strain:
Amino acid sequence according to SEP ID No. 8:
MSKIFVNPSAIRAGLADLEMAEETVDLINRNIEDNQAHLQGEPIEVDNLPEDMRQFHLGDEKLS NLGEMVRVGEGKYREDFQMDEGEDPNLLFQSYLDNVGVQIVRQMRSGERFL IWSQTVEEIIS YVTVNFPNPPGRSSEDKSTQTTGREL ETTSTLSQRESQPSKAGMVAQVASGPPSLEWSATNE EDDLSVEAEIAHQIAESFS YKFPSRSSGIFLYNFEQLEMNLDDIVKEAKNVPGVTRLAHDGSKIP LRCVLGWVALANS KFQLIVEADKLSKIMQDDLDRYTSC
Matrix Protein M (Rav M) of Flury-LEP vaccine strain:
Amino acid sequence according to SEP ID No. 9:
MNFLC IV NCRDEDTQKPSPVSAPPDGDDLWLPPPEYVPLKELTSKKNMRNFCINGEVKVCSP NGYSFRILRHILRSFDEIYSGNHRMIGLV VVIGLALSGAPVPEGMNWVYKLRRTLIFQWADSR GPLEGEELEHSQEITWDDDTEFVGLQMRVSARQCHIQGRIWCINMNSRACQLWSDMSLQTQ RSEEDKDSSLLLE
RNA Polymerase L (Rav L) of Flury-LEP vaccine strain:
Amino acid sequence according to SEP ID No. 10:
MLDPGEVYDDPIDPIESEAEPRGTPTVPNILRNSDYNLNSPLIEDSAKLMLEWLKTGNRPYRMTL TDNCSRSYKVLKDYF VDLGSLKVGGTAAQSMISLWLYGAHSESNRSRRCITDLAHFYS SSPI E LLNCTLGNRGLRIPPEGVLSCLERVDYDKAFGRYLANTYSSYLFFHVITLYMNALDWEEEKTIL ALW DLTSVDTGKDLVKF DQIWGLLVVTKDFVYSQSSNCLFDRNYTLMLKDLFLSRFNSLMIL LSPPEPRYSDDLISQLCQLYIAGDQVLSLCGNSGYEVI ILEPYVVNSLVQRAEKFRPLIHSLGDFP MFI D VNQLEGTFGPSAKRFFRVLDQFDNIHDLVFVYGCYRHWGHPYIDYRKGLSKLYDQV HIK VIDKSYQECLASDLARRILRWGFDKYSKWYLDSRFLALDHPLAPYIKTQTWPPKHIVDLV GDTWH LPITQIFEIPESMDPSEILDDKSHSFTRTRLASWLSENRGGPVPSEKVIITALS PPVNPRE FLKSIDLGGLPDDDLIIGLRPKERELKIEGRFFALMSWNLRLYFVITEKLLANYILPLFDALTMTDNL NKVF KLIDRVTGQGLLDYSRVTYAFHLDYEKWNNHQRLESTEDVFSVLDQVFGL RVFSRTH EFFQ SWIYYSDRSDLIGLREDQIYCLDMSNGPTCWNGQDGGLEGLRQ GWSLVSLLMIDRE SQTRNTRTKILAQGDNQVLCPTYMLSPGLSQEGLLYELESISRNALSIYRAIEEGASKLGLIIK EET MCSYDFLIYG TPLFRGNILVPESKRWARVSCISNDQIVNLANIMSTVSTNALTVAQHSQSLI P MRDFLLMSVQAVFHYLLFSPILKGRVYKILSAEGESFLLAMSRIIYLDPSLGGVSGMSLGRFHIRQF SDPVSEGLSFWREIWLSSHESWIHALCQEAGNPDLGERTLESFTRLLEDPTTLNIKGGASPTILL DAIRKALYDEVDKVENSEFREAILLS THRDNFILFLKSVEPLFPRFLSELFSSSFLGIPESIIGLIQNSR TIRRQFRKSLSRTLEESFYNSEIHGINRMTQTPQRVGRVWPCSSERADLLREISWGR VVGTTVP HPSEMLGLLPKSSISCTCGATGGGNPRVSVSVLPSFDQSFFSRGPLKGYLGSSTSMSTQLFHAWE VTNVHVV RAISLKESINWFINRNSNLAQTLIRNIMSLTGPDFPLEEAPVFKRTGSALHRFKSAR YSEGGYSSVCPNLLSHISVSTDTMSDLTQDGKNYDFMFQPLMLYAQTWTSELVQRDTRLRDST FHWHLRCNRCVRPIEDITLETSQIFEFPDVSKRISRMVSGAVPHFQ LPDIRLRPGDFESLSGREKS RHIGSAQGLLYSILVAIHDSGYNDGTIFPVNIYGKVSPRDYLRGLARGILIGSSICFLTRMTNINIK RPLELISGVISYILLRLDNHPSLYIMLREPSLRGEIFSIPQKIPAAYPTTMKEGNRSILCYLQHVLRYER EVITASPENDWLWIFSDFRSA MTYLTLITYQSHLLLQRVERNLSKSMRATLRQMGSLMRQVLG GHGEDTLESDDDIQRLLKDSLRRTRWVDQEVRHAARTMSGDYSPN RVSRKAGCSEWVCSA QQVAVSTSANPAPVSELDIRALS RFQNPLISGLRVVQWATGAHYKL PILDDLNVFPSLCLVV GDGSGGISRAVLNMFPDSKLVFNSLLEVNDLMASGTHPLPPSAIMSGGDDIISRVIDFDSIWE P SDLRNLATWRYFQSVQKQVNMSYDLIVCDAEVTDIASINRITLLMSDFALSIDGPLYLVF TYGT MLVNPDYKAIQHLSRAFPSVTGFITQVTSSFSSELYLRFSKRG FFRDAEYLTSSTLREMSLVLFNCS SPKSEMQRARSLNYQDLVRGFPEEIISNPYNEMIITLIDSDVESFLVHKMVDDLELQRGTLSKVAIII SIMIVFSNRVFNISKPLTDPLFYPPSDPKILRHFNICCSTMMYLSTALGDVPSFARLHDLYNRPITYY FRKQVIRGNIYLSWSWSDDTPVFKRVACNSSLSLSSHWIRLIYKIVKTTRLVGSIEDLSGEVERHL HGYNRWITLEDIRSRSSLLDYSCL
In the context of the invention additionally to the here disclosed amino acid sequences according to SEQ ID Nos. 1 -10 also amino acid sequences of different Rabies virus isolates can be used according to the invention and are incorporated herewith. These different Rabies virus isolates show preferably an identity of at least 70%, more preferably of at least 80% and most preferably of at least 90% with the amino acid sequences according to SEQ ID Nos. 1 -10.
Examples for such different Rabies virus isolates are:
Rabies virus strains according to the NCBI Accession Nos. JQ730682, AF499686, AB569299, AB8391 70, AB781935, FJ959397, AB362483, EF206720, EF20671 8, EF20671 7, EF206715, EF206714, EF206713, EF206712, EF20671 1 , EF206710, EF206709, EF206708, EF206707, EU182346, HM535790, GQ91 8139, EU877071 , EU877070, EU877069, EU1 82347, M31046, EU877068, EU877067, EF542830, AB8391 69, JQ647510, KC1 69986, JX088694, JQ730682, JN609295, JN23441 1 , HQ31 791 8, EF20671 9, EF5641 74, EU643590, JQ946087, FJ913470 HQ891318, AB645847, AB569299, AY705373, GU565704, GU565703, FJ577895, JX276550, FJ866836, FJ866835, DQ875051 , DQ875050, AB128149, AB009663, AB044824, JQ944709, EU345004, EU345003, EU345002, AB608731 , EF5641 73, JQ423952, AB618037, AB618036, AB618035, AB61 8034, AB618033, AB618032, AB085828, Ml 321 5, M21 634, AB247437, AB247436, AB247435, AB247434, AB247433, AB247432, D421 12, AB247430, and AB247431.
Furthermore in this context the coding region encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein N (RAV-N), the posphoprotein P (RAV-P), the matrix protein M (RAV-M), or the RNA polymerase L (RAV-L) of Rabies virus or a fragment, variant or derivative thereof, may be selected from any nucleic acid sequence comprising a coding region derived from any Rabies virus isolate or a fragment or variant thereof.
Particularly preferred are the wild type mRNA sequences of the coding regions of a Rabies virus vaccine strain, preferably of the Pasteur vaccine strain according to the NCBI accession No. ΜΊ3215.
Glycoprotein G (Rav G) of Pasteur vaccine strain:
Wild type mRNA sequence of the coding region according to SEQ ID No. 1 1 :
aug guu ecu cag gcu cue cug uuu gua ccc cuu cug guu uuu cca uug ugu uuu ggg aaa uuc ecu auu uac acg aua cca gac aag cuu ggu ccc ugg age ccg auu gac aua cau cac cue age ugc cca aac aau uug gua gug gag gac gaa gga ugc acc aac cug uca ggg uuc ucc uac aug gaa cuu aaa guu gga uac auc uca gcc aua aaa aug aac ggg uuc acu ugc aca ggc guu gug acg gag gcu gaa acc uac acu aac uuc guu ggu uau guc aca acc acg uuc aaa aga aag cau uuc cgc cca aca cca gau gca ugu aga gcc gcg uac aac ugg aag aug gcc ggu gac ccc aga uau gaa gag ucu cua cac aau ccg uac ecu gac uac cac ugg cuu cga acu gua aaa acc acc aag gag ucu cue guu auc aua ucu cca agu gug gca gau uug gac cca uau gac aga ucc cuu cac ucg agg guc uuc ecu ggc ggg aau ugc uca gga gua gcg gug ucu ucu acc uac ugc ucc acu aac cac gau uac acc auu ugg aug ccc gag aau ccg aga cua ggg aug ucu ugu gac auu uuu acc aau agu aga ggg aag aga gca ucc aaa ggg agu gag acu ugc ggc uuu gua gau gaa aga ggc cua uau aag ucu uua aaa gga gca ugc aaa cue aag uua ugu gga guu cua gga cuu aga cuu aug gau gga aca ugg guc gcg aug caa aca uca aau gaa acc aaa ugg ugc ecu ccc ggu cag uug gug aau uug cac gac uuu cgc uca gac gaa auu gag cac cuu guu gua gag gag uug guc aag aag aga gag gag ugu cug gau gca cua gag ucc auc aug acc acc aag uca gug agu uuc aga cgu cue agu cau uua aga aaa cuu guc ecu ggg uuu gga aaa gca uau acc aua uuc aac aag acc uug aug gaa gcc gau gcu cac uac aag uca guc aga acu ugg aau gag auc auc ecu uca aaa ggg ugu uua aga guu ggg ggg agg ugu cau ecu cau gua aac ggg gua uuu uuc aau ggu aua aua uua gga ecu gac ggc aau guc uua auc cca gag aug caa uca ucc cue cue cag caa cau aug gag uug uug gua ucc ucg guu auc ccc cuu aug cac ccc cug gca gac ccg ucu acc guu uuc aag aac ggu gac gag gcu gag gau uuu guu gaa guu cac cuu ccc gau gug cac gaa egg auc uca gga guu gac uug ggu cue ccg aac ugg ggg aag uau gua uua cug agu gca ggg gcc cug acu gcc uug aug uug aua auu uuc cug aug aca ugc ugg aga aga guc aau cga ucg gaa ecu aca caa cac aau cue aga ggg aca ggg agg gag gug uca guc acu ccc caa age ggg aag auc aua ucu uca ugg gaa uca uac aag age ggg ggu gag acc gga cug uga
Nucleoprotein N (RAV-N) of Pasteur vaccine strain:
Wild type mRNA sequence of the coding region according to SEP ID No. 12:
aug gau gcc gac aag auu gua uuc aaa guc aau aau cag gug guc ucu uug aag ecu gag auu auc gug gau caa uau gag uac aag uac ecu gcc auc aaa gau uug aaa aag ccc ugu aua acu cua gga aag gcu ccc gau uua aau aaa gca uac aag uca guu uua uca ugc aug age gcc gcc aaa cuu gau ecu gac gau gua ugu ucc uau uug gcg gcg gca aug cag uuu uuu gag ggg aca ugu ccg gaa gac ugg acc age uau gga auc gug auu gca cga aaa gga gau aag auc acc cca ggu ucu cug gug gag aua aaa cgu acu gau gua gaa ggg aau ugg gcu cug aca gga ggc aug gaa cug aca aga gac ccc acu guc ecu gag cau gcg ucc uua guc ggu cuu cue uug agu cug uau agg uug age aaa aua ucc ggg caa age acu ggu aac uau aag aca aac auu gca gac agg aua gag cag auu uuu gag aca gcc ecu uuu guu aaa auc gug gaa cac cau acu cua aug aca acu cac aaa aug ugu gcu aau ugg agu acu aua cca aac uuc aga uuu uug gcc gga acc uau gac aug uuu uuc ucc egg auu gag cau cua uau uca gca auc aga gug ggc aca guu guc acu gcu uau gaa gac ugu uca gga cug gug uca uuu acu ggg uuc aua aaa caa auc aau cue acc gcu aga gag gca aua cua uau uuc uuc cac aag aac uuu gag gaa gag aua aga aga aug uuu gag cca ggg cag gag aca gcu guu ecu cac ucu uau uuc auc cac uuc cgu uca cua ggc uug agu ggg aaa ucu ecu uau uca uca aau gcu guu ggu cac gug uuc aau cue auu cac uuu gua gga ugc uau aug ggu caa guc aga ucc cua aau gca acg guu auu gcu gca ugu gcu ecu cau gaa aug ucu guu cua ggg ggc uau cug gga gag gaa uuc uuc ggg aaa ggg aca uuu gaa aga aga uuc uuc aga gau gag aaa gaa cuu caa gaa uac gag gcg gcu gaa cug aca aag acu gac gua gca cug gca gau gau gga acu guc aac ucu gac gac gag gac uac uuc uca ggu gaa acc aga agu ccg gaa gcu guu uau acu cga auc aua aug aau gga ggu cga cug aag aga ucg cac aua egg aga uau guc uca guc agu ucc aau cau caa gcu cgu cca aac uca uuc gcc gag uuu cua aac aag aca uau ucg agu gac uca uaa
Phosphoprotein P (also named M1 ) (RAV-P) of Pasteur vaccine strain:
Wild type mRNA sequence of the coding region according to SEP ID No. 13:
aug age aag auc uuu guc aau ecu agu gcu auu aga gcc ggu cug gcc gau cuu gag aug gcu gaa gaa acu guu gau cug auc aau aga aau auc gaa gac aau cag gcu cau cue caa ggg gaa ccc aua gaa gug gac aau cue ecu gag gau aug ggg cga cuu cac cug gau gau gga aaa ucg ccc aac ecu ggu gag aug gcc aag gug gga gaa ggc aag uau cga gag gac uuu cag aug gau gaa gga gag gau ecu age cue cug uuc cag uca uac cug gac aau guu gga guc caa aua guc aga caa aua agg uca gga gag aga uuu cue aag aua ugg uca cag acc gua gaa gag auu aua ucc uau guc gcg guc aac uuu ccc aac ecu cca gga aag ucu uca gag gau aaa uca acc cag acu acc ggc cga gag cue aag aag gag aca aca ccc acu ecu ucu cag aga gaa age caa ucc ucg aaa gcc agg aug gcg gcu caa acu gcu ucu ggc ecu cca gcc cuu gaa ugg ucg gcc acc aau gaa gag gau gau cua uca gug gag gcu gag auc gcu cac cag auu gca gaa agu uuc ucc aaa aaa uau aag uuu ccc ucu cga ucc uca ggg aua cue uug uau aau uuu gag caa uug aaa aug aac cuu gau gau aua guu aaa gag gca aaa aau gua cca ggu gug acc cgu uua gcc cgu gac ggg ucc aaa cue ccc cua aga ugu gua cug gga ugg guc gcc uug gcc aac ucu aag aaa uuc cag uug uua guc gaa ucc aac aag cug agu aaa auc aug caa gau gac uug aau cgc uau aca ucu ugc uaa
Matrix protein M (also named M2) (RAV-M) of Pasteur vaccine strain:
Wild type mRNA sequence of the coding region according to SEP ID No. 14:
aug aac uuu cua cgu aag aua gug aaa aau ugc agg gac gag gac acu caa aaa ccc ucu ccc gug uca gcc ecu cug gau gac gau gac uug ugg cuu cca ccc ecu gaa uac guc ccg cua aaa gaa cuu aca age aag aag aac agg agg aac uuu ugu auc aac gga ggg guu aaa gug ugu age ccg aau ggu uac ucg uuc ggg auc cug egg cac auu cug aga uca uuc gac gag aua uau ucu ggg aau cau agg aug guc ggg uua guc aaa gua guu auu gga cug gcu uug uca gga gcu cca guc ecu gag ggc aug aac ugg gua uac aag uug agg aga acc cuu auc uuc cag ugg gcu gau ucc agg ggc ecu cuu gaa ggg gag gag uug gaa uac ucu cag gag auc acu ugg gau gau aau acu gag uuc guc gga uug caa aua aga gug agu gca aaa cag ugu cau auc egg ggc aga auc ugg ugu auc aac aug aac ucg aga gca ggu caa cua ugg ucu gac aug ucu cuu cag aca caa agg ucc gaa gag gac aaa gau ucc ucu cug cuu cua gaa uaa RNA polymerase L (RAV-L) of Pasteur vaccine strain:
Wild type mRNA sequence of the coding region according to SEQ ID No. 15:
aug cue gau ecu gga gag guc uau gau gac ecu auu gac cca auc gag uua gag gcu gaa ccc aga gga acc ccc acu guc ccc aac auc uug agg aac ucu gac uac aau cue aac ucu ecu uug aua gaa gau ecu gcu aga cua aug uua gaa ugg uua aaa aca ggg aau aga ecu uau egg aug acu cua aca gac aau ugc ucc agg ucu uuc aga guu uug aaa gau uau uuc aag aag gua gau uug ggu ucc cue aag gug ggc gga aug gcu gca cag uca aug auu ucu cue ugg uua uau ggu gec cac ucu gaa ucc aac agg age egg aga ugu aua aca gac uug gee cau uuc uau ucc aag ucg ucc ccc aua gag aag cug uua aau cue acg cua gga aau aga ggg cug aga auc ccc cca gag gga gug uua agu ugc cuu gag agg guu gau uau gau aau gca uuu gga agg uau cuu gec aac acg uau ucc ucu uac uug uuc uuc cau gua auc acc uua uac aug aac gec cua gac ugg gau gaa gaa aag acc auc cua gca uua ugg aaa gau uua acc uca gug gac auc ggg aag gac uug gua aag uuc aaa gac caa aua ugg gga cug cug auc gug aca aag gac uuu guu uac ucc caa agu ucc aau ugu cuu uuu gac aga aac uac aca cuu aug cua aaa gau cuu uuc uug ucu cgc uuc aac ucc uua aug guc uua cuu ucu ccc cca gag ccc cga uac uca gau gac uug aua ucu cag cua ugc cag cug uac auu gcu ggg gau caa guc uug ucu aug ugu gga aac ucc ggc uau gaa guc auc aaa aua uug gag cca uau guc gug aau agu uua guc cag aga gca gaa aag uuu agg ecu cue auu cau ucc uug gga gac uuu ecu gua uuu aua aaa gac aag gua agu caa cue gaa gag acg uuc ggu ucc ugu gca aga agg uuc uuu agg gcu cug gau caa uuc gac aac aua cau gac uug guu uuu gug uau ggc ugu uac agg cau ugg ggg cac cca uau aua gau uau cga aag ggu cug uca aaa cua uau gau cag guu cac auu aaa aaa gug aua gau aag ucc uac cag gag ugc uua gca age gac cua gee agg agg auc cuu aga ugg ggu uuu gau aag uac ucc aag ugg uau cug gau uca cga uuc cua gec cga gac cac ccc uug acu ecu uau auc aaa acc caa aca ugg cca ccc aaa cau auu gua gau uug gug ggg gau aca ugg cac aag cue ccg auc acg caa auc uuu gag auu ecu gaa uca aug gau cca uca gaa aua uug gau gac aaa uca cau ucu uuc acc aga acg aga cua gcu ucu ugg cug uca gaa aac cga ggg ggg ecu guu ecu age gaa aaa guu auu auc acg gee cug ucu aag ccg ecu guc aau ccc cga gag uuu cug aag ucu aua gac cue gga gga uug cca gau gaa gac uug aua auu ggc cue aag cca aag gaa egg gaa uug aag auu gaa ggu cga uuc uuu gcu cua aug uca ugg aau cua aga uug uau uuu guc auc acu gaa aaa cue uug gee aac uac auc uug cca cuu uuu gac gcg cug acu aug aca gac aac cug aac aag gug uuu aaa aag cug auc gac agg guc acc ggg caa ggg cuu cug gac uau uca agg guc aca uau gca uuu cac cug gac uau gaa aag ugg aac aac cau caa aga uua gag uca aca gag gau gua uuu ucu guc cua gau caa gug uuu gga uug aag aga gug uuu ucu aga aca cac gag uuu uuu cag aag ucc ugg auc uau uau uca gac aga uca gac cue auc ggg uua egg gag gau caa aua uac ugc uua gau gcg ucc aac ggc cca acc ugu ugg aau ggc cag gau ggc ggg cua gaa ggc uua egg cag aag ggc ugg agu cua guc age uua uug aug aua gau aga gaa ucu caa auc agg aac aca aga acc aaa gua cua gcu caa gga gac aac cag guu uua ugu ccg aca uau aug uug ucg cca ggg cua ucu caa gag ggg cue cue uau gaa uug gag age aua uea agg aau gca uuu ucg aua uac aga gcc guc gag gaa ggg gca ucu aaa cua ggg cug auc auc aag aaa gaa gag acc aug ugu agu uau gac uuc cue auc uau gga aaa acc ecu uug uuu aga ggu aac aua uug gug ecu gag ucc aaa aga ugg gcc aga guc ucu ugc guc ucu aau gac caa aua guc aac cue gcc aau aua aug ucg aca gug ucc acc aac gcg cua aca gug gca caa cac ucu caa ucu uug auc aaa ccg aug agg gau uuu cug cue aug uea gua cag gca guc uuu cac uac cug cua uuu age cca auc uua aag gga aga guu uac aag auu cug age gcu gaa ggg gag age uuu cue cua gcc aug uea agg aua auc uau cua gau ecu ucu uug gga ggg gua ucu gga aug ucc cue gga aga uuc cau aua cga cag uuc uea gac ecu guc ucu gaa ggg uua ucc uuc ugg aga gag auc ugg uua age ucc cac gag ucc ugg auu cac gcg uug ugu caa gag gcu gga aac cca gau cuu gga gag aga aca cue gag age uuc acu cgc cuu cua gaa gau ecu acc acc uua aau auc aga gga ggg gcc agu ecu acc auu cua cue aag gau gca auc aga aag gcu uua uau gac gag gug gac aag gug gag aac uea gag uuu cga gag gca auc cug uug ucc aag acc cau aga gau aau uuu aua cue uuc uua aca ucu guu gag ecu cug uuu ecu cga uuu cue agu gag cua uuc agu ucg ucu uuu uug gga auc ccc gag uea auc auu gga cug aua caa aac ucc cga acg aua aga agg cag uuu aga aag agu cue uea aaa acu uua gaa gaa ucc uuc uac aac uea gag auc cac ggg auu agu egg aug acc cag aca ecu cag agg guu ggg ggg gug ugg ecu ugc ucu uea gag agg gca gau cua cuu agg gag auc ucu ugg gga aga aaa gug gua ggc acg aca guu ecu cac ecu ucu gag aug uug ggg uua cuu ccc aag ucc ucu auu ucu ugc acu ugu gga gca aca gga gga ggc aau ecu aga guu ucu gua uea gua cue ccg ucu uuu gau cag uea uuu uuu ugc acg ggg ccc cua aag ggg uac uug ggc ucg ucc acc ucu aug ucg acc cag cua uuc cau gca ugg gaa aaa guc acu aau guu cau gug gug aag aga gcu cua ucg uua aaa gaa ucu aua aac ugg uuc auu acu aga gau ucc aac uug gcu caa acu cua auu agg aac auu gug ucu cug aca ggc ecu gau uuc ecu cua gag gag gcc ecu guu uuc aaa agg acg ggg uea gcc uug cau agg uuc aag ucu gcc aga uac age gaa gga ggg uau ucu ucu gua ugc ccg aac cue cue ucu cau auu ucu guu agu aca gac acc aug ucu gau uug acc caa gac ggg aag aac uac gau uuc aug uuc cag cca uug aug cuu uau gca cag aca ugg aca uea gag cug gua cag aga gac aca agg cua aga gac ucu acg uuu cau ugg cac cue caa ugc aac agg ugu gug aga ccc auu gac gac gug acc cug gag acc ucu cag auc uuc gag uuu ccg gau gug ucg aaa aga aua ucc aga aug guu ucu ggg gcu gug ecu cac uuc cag agg cuu ccc gau auc cgu cug aga cca gga gau uuu gaa ucu cua age ggu aga gaa aag ucu cac cau auc gga uea gcu cag ggg cue uua uac uea auc uua gug gca auu cac gac uea gga uac aau gau gga acc auc uuc ecu guc aac aua uac ggc aag guu ucc ecu aga gac uau uug aga ggg cue gca agg gga gua uug aua gga ucc ucg auu ugc uuc uug acg aga aug aca aau auc aau auu aau aga ecu cuu gaa uug auc uea ggg gua auc uea uau auu cue cug agg cua gau aac cau ccc ucc uug uac aua aug cue aga gaa ccg ucu uuu aga gaa gag aua uuu ucu auc ecu cag aaa auc ccc gcc gcu uau cca acc acu aug aaa gaa ggc aac aga uca auc uug ugu uau cue caa cau gug cua cgc uau gag cga gag gua auc acg gcg ucu cca gag aau gac ugg cua ugg auc uuu uca gac uuu aga agu gcc aaa aug acg uac cua acc cue auu acu uac cag ucu cau cuu cua cue cag agg guu gag aga aac cua ucu aag agu aug aga gau aac cug cga caa uug agu ucc uug aug agg cag gug cug ggc ggg cac gga gaa gau acc uua gag uca gac gac aac auu caa cga cua cua aaa gac ucu uua cga agg aca aga ugg gug gau caa gag gug cgc cau gca gcu aga acc aug acu gga gau uac age ccc aac aag aag gug ucc cgu aag gua gga ugu uca gaa ugg guc ugc ucu gcu caa cag guu gca guc ucu acc uca gca aac ccg gcc ecu guc ucg gag cuu gac aua agg gcc cue ucu aag agg uuc cag aac ecu uug auc ucg ggc uug aga gug guu cag ugg gca acc ggu gcu cau uau aag cuu aag ecu auu cua gau gau cue aau guu uuc cca ucu cue ugc cuu gua guu ggg gac ggg uca ggg ggg aua uca agg gca guc cue aac aug uuu cca gau gcc aag cuu gug uuc aac agu cuu uua gag gug aau gac cug aug gcu ucc gga aca cau cca cug ccU ecu uca gca auc aug agg gga gga aau gau auc guc ucc aga gug aua gau uuu gac uca auc ugg gaa aaa ccg ucc gac uug aga aac uug gcu acc ugg aaa uac uuc cag uca guc caa aag cag guc aac aug ucc uau gac cue auu auu ugc gau gca gaa guu acu gac auu gca ucu auc aac egg aua acc cug uua aug ucc gau uuu gca uug ucu aua gau gga cca cue uau uug guc uuc aaa acu uau ggg acu aug cua gua aau cca aac uac aag gcu auu caa cac cug uca aga gcg uuc ccc ucg guc aca ggg uuu auc acc caa gua acu ucg ucu uuu uca ucu gag cue uac cuu cga uuc ucc aaa cga ggg aag uuu uuc aga gau gcu gag uac uug acc ucu ucc acc cuu cga gaa aug age cuu gug uua uuc aau ugu age age ccc aag agu gag aug cag aga gcu cgu ucc uug aac uau cag gau cuu gug aga gga uuu ecu gaa gaa auc aua uca aau ecu uac aau gag aug auc aua acu cug auu gac agu gau gua gaa ucu uuu cua guc cac aag aug gug gau gau cuu gag uua cag agg gga acu cug ucu aaa gug gcu auc auu aua gcc auc aug aua guu uuc ucc aac aga guc uuc aac guu ucc aaa ccc cua acu gac ccc uug uuc uau cca ccg ucu gau ccc aaa auc cug agg cac uuc aac aua ugu ugc agu acu aug aug uau cua ucu acu gcu uua ggu gac guc ecu age uuc gca aga cuu cac gac cug uau aac aga ecu aua acu uau uac uuc aga aag caa guc auu cua ggg aac guu uau cua ucu ugg agu ugg ucc aac gac acc uca gug uuc aaa agg gua gcc ugu aau ucu age cug agu cug uca ucu cac ugg auc agg uug auu uac aag aua gug aag acu acc aga cue guu ggc age auc aag gau cua ucc gga gaa gug gaa aga cac cuu cau agg uac aac agg ugg auc acc cua gag aau auc aga ucu aga uca ucc cua cua gac uac agu ugc cug ugc auc gga uac ucc ugg aag ecu gcc cau gcu aag acu cuu gug uga
In the context of the invention additionally to the here disclosed nucleic acid sequences also nucleic acid sequences of different Rabies virus isolates are incorporated herewith. These different Rabies virus isolates show preferably an identity of at least 50%, 60%, 70%, more preferably of at least 80% and most preferably of at least 90% with the nucleic acid sequences according to SEQ ID Nos. 1 1 -1 5 or of fragments thereof.
Examples for such different Rabies virus isolates are:
Rabies virus strains according to the NCBI Accession Nos. JQ730682, AF499686, AB569299, AB8391 70, AB781935, FJ959397, AB362483, EF206720, EF20671 8, EF20671 7, EF20671 5, EF206714, EF206713, EF206712, EF20671 1 , EF206710, EF206709, EF206708, EF206707, EU182346, HM535790, GQ918139, EU877071 , EU877070, EU877069, EU1 82347, M31046, EU877068, EU877067, EF542830, AB8391 69, JQ647510, KC1 69986, JX088694, JQ730682, JN609295, JN23441 1 , HQ31 791 8, EF20671 9, EF5641 74, EU643590, JQ946087, FJ913470 HQ891318, AB645847, AB569299, AY705373, GU565704, GU565703, FJ577895, JX276550, FJ866836, FJ866835, DQ875051 , DQ875050, AB128149, AB009663, AB044824, JQ944709, EU345004, EU345003, EU345002, AB608731 , EF5641 73, JQ423952, AB61 8037, AB61 8036, AB61 8035, AB61 8034, AB61 8033, AB61 8032, AB085828, M1321 5, M21 634, AB247437, AB247436, AB247435, AB247434, AB247433, AB247432, D421 12, AB247430, and AB247431.
In a preferred embodiment, the mRNA according to the invention does not comprise a reporter gene or a marker gene. Preferably, the mRNA according to the invention does not encode, for instance, luciferase; green fluorescent protein (GFP) and its variants (such as eGFP, RFP or BFP); a-globin; hypoxanthine-guanine phosphoribosyltransferase (HGPRT); β- galactosidase; galactokinase; alkaline phosphatase; secreted embryonic alkaline phosphatase (SEAP)) or a resistance gene (such as a resistance gene against neomycin, puromycin, hygromycin and zeocin). In a preferred embodiment, the mRNA according to the invention does not encode luciferase. In another embodiment, the mRNA according to the invention does not encode GFP or a variant thereof.
In a further preferred embodiment, the mRNA according to the invention does not encode a protein (or a fragment of a protein) derived from a virus belonging to the family of Orthomyxoviridae. Preferably the mRNA does not encode a protein that is derived from an influenza virus, more preferably an influenza A virus. Preferably, the mRNA according to the invention does not encode an influenza A protein selected from the group consisting of hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), Ml , M2, NS1 , NS2 (NEP: nuclear export protein), PA, PB1 (polymerase basic 1 ), PB1 -F2 and PB2. In another preferred embodiment, the mRNA according to the invention does not encode ovalbumin (OVA) or a fragment thereof. Preferably, the mRNA according to the invention does not encode an influenza A protein or ovalbumin.
By a further embodiment, the inventive mRNA preferably comprises at least one of the following structural elements: a 5'- and/or 3'- untranslated region element (UTR element), particularly a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene or from a fragment, homolog or a variant thereof, or a 5'- and/or 3'-UTR element which may be derivable from a gene that provides a stable mRNA or from a homolog, fragment or variant thereof; a histone-stem-loop structure, preferably a histone-stem-loop in its 3' untranslated region; a 5'-CAP structure; a poly-A tail¬ or a poly(C) sequence.
In a preferred embodiment of the first aspect of the present invention the inventive mRNA comprises at least one 5'- or 3'-UTR element. In this context an UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'- or 3'-UTR of any naturally occurring gene or which is derived from a fragment, a homolog or a variant of the 5'- or 3'- UTR of a gene. Preferably the 5'- or 3'-UTR element used according to the present invention is heterologous to the coding region of the inventive mRNA sequence. Even if 5'- or 3'-UTR elements derived from naturally occurring genes are preferred, also synthetically engineered UTR elements may be used in the context of the present invention. In a particularly preferred embodiment of the first aspect of the present invention the inventive mRNA sequence comprises at least one 5'-untranslated region element (5'UTR element) which comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'UTR of a TOP gene.
It is particularly preferred that the 5'UTR element does not comprise a TOP-motif or a 5'TOP, as defined above.
In some embodiments, the nucleic acid sequence of the 5'UTR element which is derived from a 5'UTR of a TOP gene terminates at its 3'-end with a nucleotide located at position 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 upstream of the start codon (e.g. A(U T)G) of the gene or mRNA it is derived from. Thus, the 5'UTR element does not comprise any part of the protein coding region. Thus, preferably, the only protein coding part of the inventive mRNA is provided by the coding region. The nucleic acid sequence which is derived from the 5'UTR of a TOP gene is derived from a eukaryotic TOP gene, preferably a plant or animal TOP gene, more preferably a chordate TOP gene, even more preferably a vertebrate TOP gene, most preferably a mammalian TOP gene, such as a human TOP gene.
For example, the 5'UTR element is prefereably selected from 5'-UTR elements comprising or consisting of a nucleic acid sequence which is derived from a nucleic acid sequence selected from the group consisting of SEQ ID Nos. 1 -1363, SEQ ID NO. 1 395, SEQ ID NO.
1421 and SEQ ID NO. 1422 of the patent application WO201 3/143700, whose disclosure is incorporated herein by reference, from the homologs of SEQ ID Nos. 1 -1 363, SEQ ID NO.
1 395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO201 3/143700, from a variant thereof, or preferably from a corresponding RNA sequence. The term "homologs of SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO.
1422 of the patent application WO201 3/143700" refers to sequences of other species than homo sapiens, which are homologous to the sequences according to SEQ ID Nos. 1 -1 363,
SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO2013/143700.
In a preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a nucleic acid sequence extending from nucleotide position 5 (i.e. the nucleotide that is located at position 5 in the sequence) to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO201 3/143700, from the homologs of SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO201 3/143700 from a variant thereof, or a corresponding RNA sequence. It is particularly preferred that the 5' UTR element is derived from a nucleic acid sequence extending from the nucleotide position immediately 3' to the 5'TOP to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID Nos. 1 -1 363, SEQ ID NO. 1395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO2013/143700, from the homologs of SEQ ID Nos. 1 -1363, SEQ ID NO. 1 395, SEQ ID NO. 1421 and SEQ ID NO. 1422 of the patent application WO2013/143700, from a variant thereof, or a corresponding RNA sequence.
In a particularly preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal protein or from a variant of a 5'UTR of a TOP gene encoding a ribosomal protein. For example, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 1 70, 193, 244, 259, 554, 650, 675, 700, 721 , 91 3, 101 6, 1063, 1 120, 1 138, and 1284-1 360 of the patent application WO201 3/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'TOP motif. As described above, the sequence extending from position 5 to the nucleotide immediately 5' to the ATG (which is located at the 3'end of the sequences) corresponds to the 5'UTR of said sequences.
Preferably, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog or variant of a 5'UTR of a TOP gene encoding a ribosomal Large protein (RPL). For example, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 259, 1284-131 8, 1344, 1346, 1 348-1354, 1357, 1358, 1421 and 1422of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'TOP motif. In a particularly preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, or from a variant of the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, wherein preferably the 5'UTR element does not comprise the 5'TOP of said gene. Accordingly, in a particularly preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1 6 (5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract:
G GCGCTGCCTACG G AG GTG GC AGCCATCTCCTTCTCG GCATC; corresponding to SEQ ID No. 1368 of the patent application WO201 3/143700) or preferably to a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1 6 or more preferably to a corresponding RNA sequence, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR. Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein.
In some embodiments, the inventive mRNA comprises a 5'UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a vertebrate TOP gene, such as a mammalian, e.g. a human TOP gene, selected from RPSA, RPS2, RPS3, RPS3A, RPS4, RPS5, RPS6, RPS7, RPS8, RPS9, RPS10, RPS1 1 , RPS12, RPS1 3, RPS14, RPS1 5, RPS1 5A, RPS16, RPS1 7, RPS1 8, RPS19, RPS20, RPS21 , RPS23, RPS24, RPS25, RPS26, RPS27, RPS27A, RPS28, RPS29, RPS30, RPL3, RPL4, RPL5, RPL6, RPL7, RPL7A, RPL8, RPL9, RPL10, RPL10A, RPL1 1 , RPL12, RPL1 3, RPL1 3A, RPL14, RPL1 5, RPL1 7, RPL1 8, RPL1 8A, RPL1 9, RPL21 , RPL22, RPL23, RPL23A, RPL24, RPL26, RPL27, RPL27A, RPL28, RPL29, RPL30, RPL31 , RPL32, RPL34, RPL35, RPL35A, RPL36, RPL36A, RPL37, RPL37A, RPL38, RPL39, RPL40, RPL41 , RPLP0, RPLP1 , RPLP2, RPLP3, RPLP0, RPLP1 , RPLP2, EEF1 A1 , EEF1 B2, EEF1 D, EEF1 G, EEF2, EIF3E, EIF3F, EIF3H, EIF2S3, EIF3C, EIF3K, EIF3EIP, EIF4A2, PABPC1 , HNRNPA1 , TPT1 , TUBB1 , UBA52, NPM1 , ATP5G2, GNB2L1 , NME2, UQCRB, or from a homolog or variant thereof, wherein preferably the 5'UTR element does not comprise a TOP-motif or the 5'TOP of said genes, and wherein optionally the 5'UTR element starts at its 5'-end with a nucleotide located at position 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 downstream of the 5'terminal oligopyrimidine tract (TOP) and wherein further optionally the 5'UTR element which is derived from a 5'UTR of a TOP gene terminates at its 3'-end with a nucleotide located at position 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 1 0 upstream of the start codon (A(U/T)G) of the gene it is derived from.
In further particularly preferred embodiments, the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21 ), an ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 , cardiac muscle (ATP5A1 ) gene, an hydroxysteroid (1 7-beta) dehydrogenase 4 gene (HSD1 7B4), an androgen-induced 1 gene (AIG1 ), cytochrome c oxidase subunit Vic gene (COX6C), or a N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1 ) or from a variant thereof, preferably from a vertebrate ribosomal protein Large 32 gene (RPL32), a vertebrate ribosomal protein Large 35 gene (RPL35), a vertebrate ribosomal protein Large 21 gene (RPL21 ), a vertebrate ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 , cardiac muscle (ATP5A1 ) gene, a vertebrate hydroxysteroid (1 7-beta) dehydrogenase 4 gene (HSD1 7B4), a vertebrate androgen-induced 1 gene (AIG1 ), a vertebrate cytochrome c oxidase subunit Vic gene (COX6C), or a vertebrate N- acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1 ) or from a variant thereof, more preferably from a mammalian ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21 ), a mammalian ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 , cardiac muscle (ATP5A1 ) gene, a mammalian hydroxysteroid (1 7-beta) dehydrogenase 4 gene (HSD1 7B4), a mammalian androgen-induced 1 gene (AIG1 ), a mammalian cytochrome c oxidase subunit Vic gene (COX6C), or a mammalian N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1 ) or from a variant thereof, most preferably from a human ribosomal protein Large 32 gene (RPL32), a human ribosomal protein Large 35 gene (RPL35), a human ribosomal protein Large 21 gene (RPL21 ), a human ATP syn- thase, H+ transporting, mitochondrial F1 complex, alpha subunit 1 , cardiac muscle (ATP5A1 ) gene, a human hydroxysteroid (1 7-beta) dehydrogenase 4 gene (HSD1 7B4), a human androgen-induced 1 gene (AIG1 ), a human cytochrome c oxidase subunit Vic gene (COX6C), or a human N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1 ) or from a variant thereof, wherein preferably the 5'UTR element does not comprise the 5'TOP of said gene. Accordingly, in a particularly preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1 368, or SEQ ID NOs 1412-1420 of the patent application WO2013/143700, or a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 1368, or SEQ ID NOs 1412-1420 of the patent application WO2013/143700, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR. Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein.
Accordingly, in a particularly preferred embodiment, the 5'UTR element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 26 (5'-UTR of ATP5A1 lacking the 5' terminal oligopyrimidine tract: GCGGCTCGGCCATTTTGTCCCAGTCAGTCCGGAGGCTGCGGCTGCAGAAGTACCGCCT GCG-GAGTAACTGCAAAG; corresponding to SEQ ID No. 1414 of the patent application WO2013/143700) or preferably to a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID No. 26 or more preferably to a corresponding RNA sequence, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR. Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein.
In a further preferred embodiment, the inventive mRNA further comprises at least one 3'UTR element which comprises or consists of a nucleic acid sequence derived from the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
The term '3'UTR element' refers to a nucleic acid sequence which comprises or consists of a nucleic acid sequence that is derived from a 3'UTR or from a variant of a 3'UTR. A 3'UTR element in the sense of the present invention may represent the 3'UTR of an mRNA. Thus, in the sense of the present invention, preferably, a 3'UTR element may be the 3'UTR of an mRNA, preferably of an artificial mRNA, or it may be the transcription template for a 3'UTR of an mRNA. Thus, a 3'UTR element preferably is a nucleic acid sequence which corresponds to the 3'UTR of an mRNA, preferably to the 3'UTR of an artificial mRNA, such as an mRNA obtained by transcription of a genetically engineered vector construct. Preferably, the 3'UTR element fulfils the function of a 3'UTR or encodes a sequence which fulfils the function of a 3'UTR.
Preferably, the inventive mRNA comprises a 3'UTR element which may be derivable from a gene that relates to an mRNA with an enhanced half-life (that provides a stable mRNA), for example a 3'UTR element as defined and described below.
In a particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a β-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1 (I) gene, or from a variant of a 3'UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a β-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1 (I) gene according to SEQ ID No. 1 369-1 390 of the patent application WO201 3/143700 whose disclosure is incorporated herein by reference. In a particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an albumin gene, preferably a vertebrate albumin gene, more preferably a mammalian albumin gene, most preferably a human albumin gene according to SEQ ID No. 1 7
Human albumin 3'UTR SEQ ID No. 1 7:
CATCACATTT AAAAGCATCT CAGCCTACCA TGAGAATAAG AGAAAGAAAA TGAAGATCAA AAGCTTATTC ATCTGTTTTT CTTTTTCGTT GGTGTAAAGC CAACACCCTG TCTAAAAAAC ATAAATTTCT TTAATCATTT TGCCTCTTTT CTCTGTGCTT CAATTAATAA AAAATGGAAA GAATCT (corresponding to SEQ ID No: 1369 of the patent application WO201 3/143700 ).
In this context it is particularly preferred that the inventive mRNA comprises a 3'-UTR element comprising a corresponding RNA sequence derived from the nucleic acids according to SEQ ID No. 1369-1390 of the patent application WO2013/143700 or a fragment, homolog or variant thereof.
Most preferably the 3'-UTR element comprises the nucleic acid sequence derived from a fragment of the human albumin gene according to SEQ ID No. 18:
albumin7 3'UTR
CATCACATTT AAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAGAAAATGAAGATCAA TAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACAT AAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACC
T (SEQ ID No. 18 corresponding to SEQ ID No: 1 376 of the patent application WO2013/143700 )
In this context it is particularly preferred that the 3'-UTR element of the inventive mRNA comprises or consists of a corresponding RNA sequence of the nucleic acid sequence according to SEQ ID No. 18.
In another particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an a-globin gene, preferably a vertebrate a-or β-globin gene, more preferably a mammalian cc-or β-globin gene, most preferably a human a-or β-globin gene according to SEQ ID No. 1 9-21 : 3'-UTR of Homo sapiens hemoglobin, alpha 1 (HBA1 )
GCTGGAGCCTCGGTGGCCATGCTTCTTGCCCCTTGGGCCTCCCCCCAGCCCCTCCTCCC CTTCCTGCACCCGTACCCCCGTGGTCTTTGAATAAAGTCTGAGTGGGCGGC (SEQ ID No:19 corresponding to SEQ ID No. 1370 of the patent application WO201 3/143700)
3'-UTR of Homo sapiens hemoglobin, alpha 2 (HBA2)
GCTGGAGCCTCGGTAGCCGTTCCTCCTGCCCGCTGGGCCTCCCAACGGGCCCTCCTCCC CTCCTTGCACCGGCCCTTCCTGGTCTTTGAATAAAGTCTGAGTGGGCAG (SEQ ID No: 20 corresponding to SEQ ID No. 1 371 of the patent application WO2013/143700)
3'-UTR of Homo sapiens hemoglobin, beta (HBB)
GCTCGCTTTCTTGCTGTCCAATTTCTATTAAAGGTTCCTTTGTTCCCTAAGTCCAACTACTA A ACTG G G G G AT ATT ATG A AG G G CCTTG AG C ATCTG G ATTCTG CCTA ATA A A A A AC ATTT A TTTTCATTGC (SEQ ID No: 21 corresponding to SEQ ID No. 1372 of the patent application WO2013/143700)
For example, the 3'UTR element may comprise or consist of the center, a-complex-binding portion of the 3'UTR of an a-globin gene, such as of a human a-globin gene, preferably according to SEQ ID No. 22:
Center, a-complex-binding portion of the 3'UTR of an α-globin gene (also named herein as "muag")
GCCCGATGGGCCTCCCAACGGGCCCTCCTCCCCTCCTTGCACCG (SEQ ID NO. 22 corresponding to SEQ ID No. 1 393 of the patent application WO2013/143700).
In this context it is particularly preferred that the 3'-UTR element of the inventive mRNA comprises or consists of a corresponding RNA sequence of the nucleic acid sequence according to SEQ ID No. 22 or a homolog, a fragment or variant thereof.
The term 'a nucleic acid sequence which is derived from the 3'UTR of a [...] gene' preferably refers to a nucleic acid sequence which is based on the 3'UTR sequence of a [...] gene or on a part thereof, such as on the 3'UTR of an albumin gene, an α-globin gene, a β- globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1 (1) gene, preferably of an albumin gene or on a part thereof. This term includes sequences corresponding to the entire 3'UTR sequence, i.e. the full length 3'UTR sequence of a gene, and sequences corresponding to a fragment of the 3'UTR sequence of a gene, such as an albumin gene, a-globin gene, β-globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1 (1) gene, preferably of an albumin gene. The term 'a nucleic acid sequence which is derived from a variant of the 3'UTR of a [...] gene' preferably refers to a nucleic acid sequence which is based on a variant of the 3'UTR sequence of a gene, such as on a variant of the 3'UTR of an albumin gene, an a-globin gene, a β-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1 (1) gene, or on a part thereof as described above. This term includes sequences corresponding to the entire sequence of the variant of the 3'UTR of a gene, i.e. the full length variant 3'UTR sequence of a gene, and sequences corresponding to a fragment of the variant 3'UTR sequence of a gene. A fragment in this context preferably consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length variant 3'UTR, which represents at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length variant 3'UTR. Such a fragment of a variant, in the sense of the present invention, is preferably a functional fragment of a variant as described herein.
Preferably, the at least one 5'UTR element and the at least one 3'UTR element act synergistically to increase protein production from the inventive mRNA as described above.
In a particularly preferred embodiment, the inventive mRNA comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises a histone stem-loop sequence/structure. Such histone stem- loop sequences are preferably selected from histone stem-loop sequences as disclosed in WO 2012/01 9780, whose disclosure is incorporated herewith by reference.
A histone stem-loop sequence, suitable to be used within the present invention, is preferably selected from at least one of the following formulae (I) or (II): formula (I) (stem-loop sequence without stem bordering elements):
[N0-2GN3-5] [No-4(U/T)No-4] [N3-5CN0-2]
Y > J V ' steml loop stem2 formula (II) (stem-loop sequence with stem bordering elements):
N1-6 [N0-2GN3-5] [No-4(U/T)No-4] [N3-5CN0-2] Ni-6 steml steml loop stem2 stem2 bordering element bordering element wherein: steml or stem2 bordering elements N1-6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof;
steml [N0-2GN3-5] is reverse complementary or partial ly reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1, more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof, and wherein G is guanosine or an analogue thereof, and may be optionally replaced by a cytidine or an analogue thereof, provided that its complementary nucleotide cytidine in stem2 is replaced by guanosine; loop sequence [N0-4(U/T)N0-4] is located between elements steml and stem2, and is a consecutive sequence of 3 to 5 nucleotides, more preferably of 4 nucleotides; wherein each N0-4 is independent from another a consecutive sequence of 0 to 4, preferably of 1 to 3, more preferably of 1 to 2 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; and
wherein U/T represents uridine, or optionally thymidine; stem2 [N3-5CN0-2] is reverse complementary or partially reverse complementary with element steml , and is a consecutive sequence between of 5 to 7 nucleotides; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T,
G and C or a nucleotide analogue thereof; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G or C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleoside guanosine in stemi is replaced by cytidine; wherein
stemi and stem2 are capable of base pairing with each other forming a reverse complementary sequence, wherein base pairing may occur between stemi and stem2, e.g. by Watson-Crick base pairing of nucleotides A and U T or G and C or by non-Watson-Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between stemi and stem2, on the basis that one ore more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
According to a further preferred embodiment of the first inventive aspect, the inventive mRNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (la) or (11a): formula (la) (stem-loop sequence without stem bordering elements):
[N0-1 G N3-5] [Ni-3(U/T)No-2] [N3-5CN0-1]
— ' Y J ' +—
stemi loop stem2 formula (I la) (stem-loop sequence with stem bordering elements):
N2-5 [N0-1 G N3-5] [Ni-3(U/T)No-2] [ N3-5CN0-1 ] N2-5
- ' y v~
steml steml loop stem2 stem2
bordering element bordering element wherein: N, C, G, T and U are as defined above.
According to a further more particularly preferred embodiment of the first aspect, the inventive mRNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (lb) or (Mb): formula (lb) (stem-loop sequence without stem bordering elements):
[N1GN4] [N2(U T)Ni ] [N4CN1]
stem 1 loop stem2 formula (lib) (stem-loop sequence with stem bordering elements): N4-5 [Ni GNJ [N2(U T)Ni ] [N4CN1] N4-5 steml steml loop stem2 stem2
bordering element bordering element
wherein:
N, C, G, T and U are as defined above. A particular preferred histone stem-loop sequence is the sequence according to SEQ ID NO: 23 CAAAGGCTCTTTTCAGAGCCACCA or more preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 27 (CAAAGGCUCUUUUCAGAGCCACCA SEQ ID NO: 27).
In a particular preferred embodiment of the first aspect of the present invention the inventive mRNA comprises additionally to the coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, a poly(A) sequence, also called poly-A-tail, preferably at the 3'-terminus of the inventive mRNA. When present, such a poly(A) sequence comprises a sequence of about 25 to about 400 adenosine nucleotides, preferably a sequence of about 50 to about 400 adenosine nucleotides, more preferably a sequence of about 50 to about 300 adenosine nucleotides, even more preferably a sequence of about 50 to about 250 adenosine nucleotides, most preferably a sequence of about 60 to about 250 adenosine nucleotides. In this context the term "about" refers to a deviation of ± 10% of the value(s) it is attached to. This poly(A) sequence is preferably located 3' of the coding region comprised in the inventive mRNA according to the first aspect of the present invention.
According to a further preferred embodiment the inventive mRNA can be modified by a sequence of at least 1 0 cytosines, preferably at least 20 cytosines, more preferably at least 30 cytosines (so-called "poly(C) sequence"). Particularly, the mRNA may contain a poly(C) sequence of typically about 1 0 to 200 cytosine nucleotides, preferably about 10 to 100 cytosine nucleotides, more preferably about 1 0 to 70 cytosine nucleotides or even more preferably about 20 to 50 or even 20 to 30 cytosine nucleotides. This poly(C) sequence is preferably located 3' of the coding region, more preferably 3' of an optional poly(A) sequence comprised in the inventive mRNA according to the first aspect of the present invention.
In this context the inventive mRNA sequence may comprise in a specific embodiment: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c.) a poly(A) sequence preferably comprising 64 adenosines; and d.) optionally, a poly(C) sequence, preferably comprising 30 cytosines. In a particularly preferred embodiment of the first aspect of the present invention the inventive mRNA comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c. ) a poly(A) sequence preferably comprising 64 adenosines; d. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and e. ) a histone-stem-loop, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 23.
In a further particularly preferred embodiment of the first aspect of the present invention the inventive mRNA comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c. ) optionally, a 3'-UTR element derived from an alpha globin gene, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22, a homolog, a fragment, or a variant thereof; d. ) a poly(A) sequence preferably comprising 64 adenosines; e. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and f.) a histone-stem-loop, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 23. In another particular preferred embodiment the inventive mRNA encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof, comprises preferably in 5'- to 3'-direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) optionally, a 5'-UTR element derived from a TOP gene, preferably derived from the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6 , a homolog, a fragment, or a variant thereof; c. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; d. ) optionally, a 3'UTR element derived of a gene providing a stable mRNA, preferably derived from the corresponding RNA sequence of a nucleic acid sequence according to SEQ ID NO. 18, a homolog, a fragment, or a variant thereof; e. ) a poly(A) sequence preferably comprising 64 adenosines; f. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and g. ) a histone-stem-loop, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 23.
The coding region might encode at least partially one of the amino acid sequences according to SEQ ID Nos. 1 -10 or fragments, variants or derivatives thereof. Furthermore the coding region of the inventive mRNA may encode a combination of at least two of these amino acid sequences or a combination of fragments, variants or derivatives thereof.
Additionally the coding region might be or might comprise at least partially one of the sequences according to SEQ ID No. 1 1 to SEQ ID No. 1 5, or fragments, homologs or variants thereof. Furthermore, the mRNA might comprise a combination of at least two of these sequences or a combination of fragments, homologs or variants thereof.
For further improvement of the resistance to e.g. in vivo degradation (e.g. by an exo- or endo-nuclease), the inventive mRNA may be provided as a stabilized nucleic acid, e.g. in the form of a modified nucleic acid. According to a further embodiment of the invention it is therefore preferred that the inventive mRNA is stabilized, preferably by backbone modifications, sugar modifications and/or base modifications, more preferred stabilized by modification of the G/C-content. All of these modifications may be introduced into the inventive mRNA without impairing the mRNA's function to be translated into the antigenic function derived from the Rabies virus peptide or protein.
A backbone modification in the context of the present invention is preferably a modification in which phosphates of the backbone of the nucleotides contained in the inventive mRNA are chemically modified, e.g. anionic i nternucleoside li nkage, N3'->P5' modifications, replacement of non-bridging oxygen atoms by boranes, neutral internucleoside linkage, amide linkage of the nucleosides, methylene(methylimino) linkages, formacetal and thioformacetal linkages, introduction of sulfonyl groups, or the like. A sugar modification in the context of the present invention is preferably a chemical modification of the sugar of the nucleotides of the inventive mRNA, e.g. methylation of the ribose residue or the like.
According to another embodiment, the inventive mRNA may be modified and thus stabilized by modifying the G (guanosine)/C (cytosine) content of the mRNA, preferably of the coding region thereof.
Therein, the G/C content of the inventive mRNA, preferably of the coding region, is particularly increased compared to the G/C content of the coding region of its particular wild type coding sequence, i.e. the unmodified mRNA. However, the encoded amino acid sequence of the inventive mRNA is preferably not modified compared to the coded amino acid sequence of the particular wild type/unmodified mRNA.
The modification of the G/C-content of the inventive mRNA is based on the fact that RNA sequences having an increased G (guanosine)/C (cytosine) content are more stable than RNA sequences having an increased A (adenosine)/U (uracil) content. The codons of a coding sequence or a whole RNA might therefore be varied compared to the wild type coding sequence or mRNA, such that they include an increased amount of G/C nucleotides whi le the translated amino acid sequence is retained. In respect to the fact that several codons code for one and the same amino acid (so-called degeneration of the genetic code), the most favourable codons for the stabi lity can be determined (so-called alternative codon usage). Preferably, the G/C content of the coding region of the inventive mRNA according to the invention is increased by at least 7%, more preferably by at least 1 5%, particularly preferably by at least 20%, compared to the G/C content of the coded region of the wi ld type RNA. According to a specific embodiment at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, more preferably at least 70 %, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for a protein or peptide as defined herein or its fragment or variant thereof or the whole sequence of the wild type mRNA sequence or coding sequence are substituted, thereby increasing the G/C content of said sequence. In this context, it is particularly preferable to increase the G/C content of the inventive mRNA to the maximum (i.e. 1 00% of the substitutable codons), in particular in the coding region, compared to the wild type sequence.
According to a further preferred embodiment of the invention, the inventive mRNA is optimized for translation, preferably optimized for translation by replacing codons for less frequent tRNAs of a given amino acid by codons for more frequently occurring tRNAs of the respective amino acid. This is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, if so-called "less frequent codons" are present in the inventive mRNA to an increased extent, the corresponding modified RNA is translated to a significantly poorer degree than in the case where codons coding for more frequent tRNAs are present. Preferably, the coding region of the inventive mRNA is modified compared to the corresponding region of the wild type RNA or coding sequence such that at least one codon of the wild type sequence which codes for a tRNA which is relatively rare or less frequent in the cell is exchanged for a codon which codes for a tRNA which is more or most frequent in the cell and carries the same amino acid as the relatively rare or less frequent tRNA. By this modification, the sequences of the inventive mRNA can be modified such that codons for which more frequently occurring tRNAs are available are inserted. In other words, according to the invention, by this modification all codons of the wild type sequence which code for a tRNA which is relatively rare in the cell can in each case be exchanged for a codon which codes for a respective tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA. Furthermore, it is particularly preferable to link the sequential G/C content which is increased, in particular maximized, in the inventive mRNA with the "frequent" codons without modifying the amino acid sequence of the protein encoded by the coding region of the inventive mRNA or of the coding region. This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) inventive mRNA.
Substitutions, additions or eliminations of bases are preferably carried out using a DNA matrix for preparation of the nucleic acid molecule by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation. In such a process, for preparation of the at least one RNA of the inventive combination vaccine as defined herein a corresponding DNA molecule may be transcribed in vitro. This DNA matrix preferably comprises a suitable promoter, e.g. a T7 or SP6 promoter, for in vitro transcription, which is followed by the desired nucleotide sequence for the at least one RNA to be prepared and a termination signal for in vitro transcription. The DNA molecule, which forms the matrix of the at least one RNA of interest, may be prepared by fermentative proliferation and subsequent isolation as part of a plasmid which can be replicated in bacteria. Plasmids which may be mentioned as suitable for the present invention are e.g. the plasmids pT7Ts (GenBank accession number U26404; Lai et a/., Development 1995, 121 : 2349 to 2360), pGEM® series, e.g. pGEM®-1 (GenBank accession number X65300; from Promega) and pSP64 (GenBank accession number X65327); cf. also Mezei and Starts, Purification of PCR Products, in: Griffin and Griffin (ed.), PCR Technology: Current Innovation, CRC Press, Boca Raton, FL, 2001 . In a particularly preferred embodiment, the inventive mRNA sequence according to the first aspect of the present invention comprises, preferably in 5'- to 3'- direction:
a) a 5'-CAP structure, as defined herein, preferably m7GpppN;
b) a coding region, preferably with an increased or even maximized G/C content compared with the G/C content of the coding region of the wild type mRNA, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), or the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof; c) a 3'-UTR element as defined herein, preferably derived of a gene providing a stable mRNA, most preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22, or a homolog, a fragment or variant thereof;
d) a poly(A) sequence, preferably consisting of 64 adenosines
e) optionally a poly(C) sequence, preferably consisting of 30 cytosines.
f) at least one histone stem-loop sequence, preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 23.
Most preferably, the inventive mRNA sequence of that specific embodiment comprises the sequence modifications as shown in Fig. 1 (SEQ ID NO. 24).
In a further particularly preferred embodiment, the inventive mRNA sequence according to the first aspect of the present invention comprises preferably in 5' to 3' direction: a) a 5'-CAP structure, as defined herein, preferably m7GpppN;
b) a 5'-UTR element as defined herein, preferably a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, preferably the 5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract according to SEQ ID NO. 1 6 or the corresponding RNA sequence; or a fragment, homolog or variant thereof;
c) a coding region, preferably with an increased or even maximized G/C content compared with the G/C content of the coding region of the wild type mRNA, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), the nucleoprotein (RAV-N), the posphoprotein (RAV-P), the matrix protein (RAV-M), and the RNA polymerase (RAV-L) of Rabies virus or a fragment, variant or derivative thereof;
d) a 3'-UTR element, preferably the 3'-UTR element of human albumin according to SEQ ID No. 18 or the corresponding RNA, or a homolog, a fragment or a variant thereof; e) a poly(A) sequence, preferably consisting of 64 adenosines
f) optionally a poly(C) sequence, preferably consisting of 30 cytosines.
g) at least one histone stem-loop sequence, preferably the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 23. Most preferably, the inventive mRNA of that specific embodiment comprises the sequence modifications as shown in Fig. 2 (SEQ ID NO. 25).
In an even more particularly preferred embodiment the inventive mRNA comprises or consists of the sequences shown in Fig. 1 or 2 according to SEQ ID Nos. 24 and 25.
In further specific embodiments, the mRNA according to the invention may further comprise an internal ribosome entry site (IRES) sequence or IRES-motif, which may separate several open reading frames, for example if the inventive mRNA encodes for two or more antigenic peptides or proteins. An IRES-sequence may be particularly helpful if the mRNA is a bi- or multicistronic mRNA.
Additionally, the inventive mRNA may be prepared using any method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as in vitro methods, such as in vitro transcription reactions. According to one embodiment of the present invention the mRNA comprising a coding region, encoding at least one antigenic peptide or protein of Rabies virus or a fragment, variant or derivative thereof may be administered naked without being associated with any further vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the inventive mRNA or of further comprised nucleic acid.
In a preferred embodiment, the inventive mRNA may be formulated together with a cationic or polycationic compound and/or with a polymeric carrier. Accordingly, in a further embodiment of the invention it is preferred that the inventive mRNA or any other nucleic acid comprised in the inventive pharmaceutical composition or vaccine is associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w) of mRNA or nucleic acid to cationic or polycationic compound and/or with a polymeric carrier; or optionally in a nitrogen/phosphate ratio of mRNA or nucleic acid to cationic or polycationic compound and/or polymeric carrier in the range of about 0.1 -10, preferably in a range of about 0.3-4 or 0.3-1 , and most preferably in a range of about 0.5-1 or 0.7-1 , and even most preferably in a range of about 0.3-0.9 or 0.5-0.9.
Thereby, the inventive mRNA or any other nucleic acid comprised in the inventive pharmaceutical composition or vaccine can also be associated with a vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the inventive mRNA or of optionally comprised further included nucleic acids.
Cationic or polycationic compounds, being particularly preferred agents in this context include protamine, nucleoline, spermine or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs), PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1 , L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, plsl, FGF, Lactoferrin, Transportan, Buforin-2, Bac715-24, SynB, SynB(1 ), pVEC, hCT-derived peptides, SAP, or histories.
In this context protamine is particularly preferred.
Additionally, preferred cationic or polycationic proteins or peptides may be selected from the following proteins or peptides having the following total formula (III):
(Arg)i;(Lys)m;(His)n;(Orn)0;(Xaa)x, (formula (III)) wherein I + m + n +o + x = 8-1 5, and I, m, n or o independently of each other may be any number selected from 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 1 3, 14 or 15, provided that the overall content of Arg, Lys, His and Orn represents at least 50% of all amino acids of the oligopeptide; and Xaa may be any amino acid selected from native (= naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x may be any number selected from 0, 1 , 2, 3 or 4, provided, that the overall content of Xaa does not exceed 50 % of all amino acids of the oligopeptide. Particularly preferred cationic peptides in this context are e.g. Argy, Arg8, Arg9, H3R9, R9H3, H3R9H3, YSSR9SSY, (RKH)4, Y(RKH)2R, etc. In this context the disclosure of WO 2009/030481 is incorporated herewith by reference.
Further preferred cationic or polycationic compounds, which can be used as transfection or complexation agent may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g. DOTMA: [1 -(2,3- sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Choi, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol- amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DIMRI: Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3- (trimethylammonio)propane, DC-6-14: O,O-ditetradecanoyl-N-(a- trimethylammonioacetyl)diethanolamine chloride, CLIP1 : rac-[(2,3-dioctadecyloxypropyl)(2- hydroxyethyl)]-dimethylammonium chloride, CLIP6: rac-[2(2,3-dihexadecyloxypropyl- oxymethyloxy)ethyl]trimethylammonium, CLIP9: rac-[2(2,3-dihexadecyloxypropyl- oxysuccinyloxy)ethyl]-trimethylammonium, oligofectamine, or cationic or polycationic polymers, e.g. modified polyaminoacids, such as β-aminoacid-polymers or reversed polyamides, etc., modified polyethylenes, such as PVP (poly(N-ethyl-4-vinylpyridinium bromide)), etc., modified acrylates, such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc., modified amidoamines such as pAMAM (poly(amidoamine)), etc., modified polybetaaminoester (PBAE), such as diamine end modified 1 ,4 butanediol diacrylate-co-5-amino-1 -pentanol polymers, etc., dendrimers, such as polypropylamine dendrimers or pAMAM based dendrimers, etc., polyimine(s), such as PEI: poly(ethyleneimine), poly(propyleneimine), etc., polyallylamine, sugar backbone based polymers, such as cyclodextrin based polymers, dextran based polymers, chitosan, etc., si Ian backbone based polymers, such as PMOXA-PDMS copolymers, etc., blockpolymers consisting of a combination of one or more cationic blocks (e.g. selected from a cationic polymer as mentioned above) and of one or more hydrophilic or hydrophobic blocks (e.g. polyethyleneglycole); etc.
A polymeric carrier used according to the invention might be a polymeric carrier formed by disulfide-crosslinked cationic components. The disulfide-crosslinked cationic components may be the same or different from each other. The polymeric carrier can also contain further components. It is also particularly preferred that the polymeric carrier used according to the present invention comprises mixtures of cationic peptides, proteins or polymers and optionally further components as defined herein, which are crosslinked by disulfide bonds as described herein. In this context the disclosure of WO 2012/013326 is incorporated herewith by reference.
In this context the cationic components, which form basis for the polymeric carrier by disulfide-crosslinkage, are typically selected from any suitable cationic or polycationic peptide, protein or polymer suitable for this purpose, particular any cationic or polycationic peptide, protein or polymer capable to complex an mRNA or a nucleic acid as defined according to the present invention, and thereby preferably condensing the mRNA or the nucleic acid. The cationic or polycationic peptide, protein or polymer, is preferably a linear molecule, however, branched cationic or polycationic peptides, proteins or polymers may also be used.
Every disulfide-crosslinking cationic or polycationic protein, peptide or polymer of the polymeric carrier, which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine contains at least one -SH moiety, most preferably at least one cysteine residue or any further chemical group exhibiting an -SH moiety, capable to form a disulfide linkage upon condensation with at least one further cationic or polycationic protein, peptide or polymer as cationic component of the polymeric carrier as mentioned herein. As defined above, the polymeric carrier, which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine may be formed by disulfide-crosslinked cationic (or polycationic) components.
Preferably, such cationic or polycationic peptides or proteins or polymers of the polymeric carrier, which comprise or are additionally modified to comprise at least one -SH moiety, are selected from, proteins, peptides and polymers as defined above for complexation agent.
In a further particular embodiment, the polymeric carrier which may be used to complex the inventive mRNA or any further nucleic acid comprised in the inventive pharmaceutical composition or vaccine may be selected from a polymeric carrier molecule according to generic formula (IV):
L-pi-S-[S-P2-S]n-S-P3-L formula (IV) wherein,
P1 and P3 are different or identical to each other and represent a linear or branched hydrophilic polymer chain, each P' and P3 exhibiting at least one -SH-moiety, capable to form a disulfide linkage upon condensation with component P2, or alternatively with (AA), (AA)X, or [(AA)X]2 if such components are used as a linker between P1 and P2 or P3 and P2) and/or with further components (e.g. (AA), (AA)X, [(AA)x] z or L), the linear or branched hydrophilic polymer chain selected independent from each other from polyethylene glycol (PEG), poly-/V-(2- hydroxypropyl)methacrylamide, poly-2-(methacryloyloxy)ethyl phosphorylcholines, poly(hydroxyalkyl L-asparagine), poly(2-
(methacryloyloxy)ethyl phosphorylcholine), hydroxyethylstarch or poly(hydroxyalkyl L-glutamine), wherein the hydrophilic polymer chain exhibits a molecular weight of about 1 kDa to about 100 kDa, preferably of about 2 kDa to about 25 kDa; or more preferably of about 2 kDa to about 1 0 kDa, e.g. about 5 kDa to about 25 kDa or 5 kDa to about 10 kDa;
P2 is a cationic or polycationic peptide or protein, e.g. as defined above for the polymeric carrier formed by disulfide-crosslinked cationic components, and preferably having a length of about 3 to about 1 00 amino acids, more preferably having a length of about 3 to about 50 amino acids, even more preferably having a length of about 3 to about 25 amino acids, e.g. a length of about 3 to 10, 5 to 1 5, 10 to 20 or 1 5 to 25 amino acids, more preferably a length of about 5 to about 20 and even more preferably a length of about 1 0 to about 20; or is a cationic or polycationic polymer, e.g. as defined above for the polymeric carrier formed by disulfide-crosslinked cationic components, typically having a molecular weight of about 0.5 kDa to about 30 kDa, including a molecular weight of about 1 kDa to about 20 kDa, even more preferably of about 1 .5 kDa to about 10 kDa, or having a molecular weight of about 0.5 kDa to about 100 kDa, including a molecular weight of about 10 kDa to about 50 kDa, even more preferably of about 10 kDa to about 30 kDa; each P2 exhibiting at least two -SH-moieties, capable to form a disulfide linkage upon condensation with further components P2 or component(s) P1 and/or P3 or alternatively with further components (e.g. (AA), (AA)X, or [(AA)X]Z); is a (reversible) disulfide bond (the brackets are omitted for better readability), wherein S preferably represents sulphur or a -SH carrying moiety, which has formed a (reversible) disulfide bond. The (reversible) disulfide bond is preferably formed by condensation of -SH-moieties of either components P1 and P2, P2 and P2, or P2 and P3, or optionally of further components as defined herein (e.g. L, (AA), (AA)x, [(AA)x]z, etc); The -SH-moiety may be part of the structure of these components or added by a modification as defined below; is an optional ligand, which may be present or not, and may be selected independent from the other from RGD, Transferrin, Folate, a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide, (e.g. TAT or KALA), a ligand of a receptor (e.g. cytokines, hormones, growth factors etc), small molecules (e.g. carbohydrates like mannose or galactose or synthetic ligands), small molecule agonists, inhibitors or antagonists of receptors (e.g. RGD peptidomimetic analogues), or any further protein as defined herein, etc.; is an integer, typically selected from a range of about 1 to 50, preferably from a range of about 1 , 2 or 3 to 30, more preferably from a range of about 1 , 2, 3, 4, or 5 to 25, or a range of about 1 , 2, 3, 4, or 5 to 20, or a range of about 1 , 2, 3, 4, or 5 to 1 5, or a range of about 1 , 2, 3, 4, or 5 to 10, including e.g. a range of about 4 to 9, 4 to 10, 3 to 20, 4 to 20, 5 to 20, or 10 to 20, or a range of about 3 to 1 5, 4 to 1 5, 5 to 1 5, or 1 0 to 1 5, or a range of about 6 to 1 1 or 7 to 1 0. Most preferably, n is in a range of about 1 , 2, 3, 4, or 5 to 10, more preferably in a range of about 1 , 2, 3, or 4 to 9, in a range of about 1 , 2, 3, or 4 to 8, or i n a range of about 1 , 2, or 3 to 7.
In this context the disclosure of WO 201 1 /026641 is incorporated herewith by reference. Each of hydrophilic polymers P1 and P3 typical ly exhibits at least one -SH-moiety, wherein the at least one -SH-moiety is capable to form a disulfide linkage upon reaction with component P2 or with component (AA) or (AA)X, if used as linker between P1 and P2 or P3 and P2 as defined below and optionally with a further component, e.g. L and/or (AA) or (AA)X, e.g. if two or more -SH-moieties are contained. The followi ng subformulae "P1-S-S-P2" and "P2-S-S-P3" within generic formula (V) above (the brackets are omitted for better readability), wherein any of S, P' and P3 are as defined herein, typical ly represent a situation, wherein one-SH-moiety of hydrophilic polymers P1 and P3 was condensed with one -SH- moiety of component P2 of generic formula (V) above, wherein both sulphurs of these -SH- moieties form a disulfide bond -S-S- as defined herein in formula (V). These -SH-moieties are typically provided by each of the hydrophilic polymers P1 and P3, e.g. via an internal cysteine or any further (modified) amino acid or compound which carries a -SH moiety. Accordingly, the subformulae "P1-S-S-P2" and "P2-S-S-P3" may also be written as "P'-Cys- Cys-P2" and "P2-Cys-Cys-P3", if the -SH- moiety is provided by a cysteine, wherein the term Cys-Cys represents two cysteines coupled via a disulfide bond, not via a peptide bond. In this case, the term "-S-S-" in these formulae may also be written as "-S-Cys", as "-Cys-S" or as "-Cys-Cys-". In this context, the term "-Cys-Cys-" does not represent a peptide bond but a linkage of two cysteines via their -SH-moieties to form a disulfide bond. Accordingly, the term "-Cys-Cys-" also may be understood generally as "-(Cys-S)-(S-Cys)-", wherein in this specific case S indicates the sulphur of the -SH-moiety of cysteine. Likewise, the terms "-S- Cys" and "-Cys-S" indicate a disulfide bond between a -SH containing moiety and a cysteine, which may also be written as "-S-(S-Cys)" and "-(Cys-S)-S". Alternatively, the hydrophilic polymers P1 and P3 may be modified with a -SH moiety, preferably via a chemical reaction with a compound carrying a -SH moiety, such that each of the hydrophilic polymers P1 and P3 carries at least one such -SH moiety. Such a compound carrying a -SH moiety may be e.g. an (additional) cysteine or any further (modified) amino acid, which carries a -SH moiety. Such a compound may also be any non-amino compound or moiety, which contains or al lows to introduce a -SH moiety into hydrophilic polymers P1 and P3 as defined herein. Such non-amino compounds may be attached to the hydrophilic polymers P1 and P3 of formula (VI) of the polymeric carrier according to the present invention via chemical reactions or binding of compounds, e.g. by binding of a 3-thio propionic acid or thioimolane, by amide formation (e.g. carboxylic acids, sulphonic acids, amines, etc), by Michael addition (e.g maleinimide moieties, ,β unsatured carbonyls, etc), by click chemistry (e.g. azides or alkines), by alkene/alkine methatesis (e.g. alkenes or alkines), imine or hydrozone formation (aldehydes or ketons, hydrazins, hydroxylamins, amines), complexation reactions (avidin, biotin, protein G) or components which allow Sn- type substitution reactions (e.g halogenalkans, thiols, alcohols, amines, hydrazines, hydrazides, sulphonic acid esters, oxyphosphonium salts) or other chemical moieties which can be utilized in the attachment of further components. A particularly preferred PEG derivate in this context is alpha-Methoxy-omega-mercapto poly(ethylene glycol). In each case, the SH-moiety, e.g. of a cysteine or of any further (modified) amino acid or compound, may be present at the terminal ends or internally at any position of hydrophilic polymers P1 and P3. As defined herein, each of hydrophilic polymers P1 and P3 typically exhibits at least one -SH-moiety preferably at one terminal end, but may also contain two or even more - SH-moieties, which may be used to additionally attach further components as defined herein, preferably further functional peptides or proteins e.g. a ligand, an amino acid component (AA) or (AA)X, antibodies, cell penetrating peptides or enhancer peptides (e.g. TAT, KALA), etc.
In this context it is particularly preferred that the inventive mRNA is complexed at least partially with a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides. In this context the disclosure of WO 2010/037539 and WO 2012/1 1351 3 is incorporated herewith by reference. Partially means that only a part of the inventive mRNA is complexed with a cationic compound and that the rest of the inventive mRNA is (comprised in the inventive pharmaceutical compostion or vaccine) in uncomplexed form ("free"). Preferably the ratio of complexed mRNA to: free mRNA (in the inventive pharmaceutical composition or vaccine) is selected from a range of about 5:1 (w/w) to about 1 :10 (w/w), more preferably from a range of about 4:1 (w/w) to about 1 :8 (w/w), even more preferably from a range of about 3:1 (w/w) to about 1 :5 (w/w) or 1 :3 (w/w), and most preferably the ratio of complexed mRNA to free mRNA in the inventive pharmaceutical composition or vaccine is selected from a ratio of about 1 :1 (w/w). The complexed mRNA in the inventive pharmaceutical composition or vaccine, is preferably prepared according to a first step by complexing the inventive mRNA with a cationic or polycationic compound and/or with a polymeric carrier, preferably as defined herein, in a specific ratio to form a stable complex. In this context, it is highly preferable, that no free cationic or polycationic compound or polymeric carrier or only a negligibly small amount thereof remains in the component of the complexed mRNA after complexing the mRNA. Accordingly, the ratio of the mRNA and the cationic or polycationic compound and/or the polymeric carrier in the component of the complexed mRNA is typically selected in a range that the mRNA is entirely complexed and no free cationic or polycationic compound or polymeric carrier or only a negligibly small amount thereof remains in the composition.
Preferably the ratio of the mRNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, is selected from a range of about 6:1 (w/w) to about 0,25:1 (w/w), more preferably from about 5:1 (w/w) to about 0,5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w). Alternatively, the ratio of the mRNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, in the component of the complexed mRNA, may also be calculated on the basis of the nitrogen/phosphate ratio (N/P-ratio) of the entire complex. In the context of the present invention, an N/P-ratio is preferably in the range of about 0.1 -1 0, preferably in a range of about 0.3-4 and most preferably in a range of about 0.5-2 or 0.7-2 regarding the ratio of mRNA: cationic or polycationic compound and/or polymeric carrier, preferably as defined herein, in the complex, and most preferably in a range of about 0.7- 1 ,5, 0.5-1 or 0.7-1 , and even most preferably in a range of about 0.3-0.9 or 0.5-0.9., preferably provided that the cationic or polycationic compound in the complex is a cationic or polycationic cationic or polycationic protein or peptide and/or the polymeric carrier as defined above. In this specific embodiment the complexed mRNA is also emcompassed in the term "adjuvant component".
In a further aspect the invention provides for a composition comprising a plurality or more than one, preferably 2 to 10, more preferably 2 to 5, most preferably 2 to 4 of the inventive mRNA sequences as defined herein. These inventive compositions comprise more than one inventive mRNA sequences, preferably encoding different peptides or proteins which comprise preferably different pathogenic antigens or fragments, variants or derivatives thereof. Particularly preferred in this context is that at least one mRNA sequence encodes at least one antigenic peptide or protein derived from glycoprotein G (RAV-G) of Rabies virus and that at least one mRNA sequence encodes at least one antigenic peptide or protein derived from another antigen of Rabies virus, particularly of nucleoprotein N (RAV-N). Accordingly, in a further particular preferred aspect, the present invention also provides a pharmaceutical composition, comprising at least one inventive mRNA sequence as defined herein or an inventive composition comprising a plurality of inventive mRNA sequences as defined herein and optional ly a pharmaceutically acceptable carrier and/or vehicle. As a first ingredient, the inventive pharmaceutical composition comprises at least one inventive mRNA sequence as defined herein.
As a second ingredient the inventive pharmaceutical composition may optional comprise at least one additional pharmaceutical ly active component. A pharmaceutically active component in this connection is a compound that has a therapeutic effect to heal, ameliorate or prevent a particular indication or disease as mentioned herein, preferably rabies. Such compounds include, without implying any limitation, peptides or proteins, preferably as defined herein, nucleic acids, preferably as defi ned herein, (therapeutically active) low molecular weight organic or inorganic compounds (molecular weight less than 5000, preferably less than 1000), sugars, antigens or antibodies, preferably as defined herein, therapeutic agents already known in the prior art, antigenic cells, antigenic cel lular fragments, cel lular fractions; cell wall components (e.g. polysaccharides), modified, attenuated or de-activated (e.g. chemically or by irradiation) pathogens (virus, bacteria etc.), adjuvants, preferably as defined herein, etc. Particularly preferred i n this context are rabies vaccines, e.g. "Tol lwutimpfstoff (HDC) inaktiviert" or Rabipur, or rabies immune globulines, e.g. Merieux P or Berirab.
The inventive pharmaceutical composition may be administered orally, parenteral ly, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques. Particularly preferred is intradermal and intramuscular injection. Sterile injectable forms of the inventive pharmaceutical compositions may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
Preferably, the inventive vaccine may be administered by conventional needle injection or needle-free jet injection. In a preferred embodiment the inventive pharmaceutical composition may be administered by jet injection as defined herein, preferably intramuscularly or intradermal ly, more preferably intradermally. According to a specific embodiment, the inventive pharmaceutical composition may comprise an adjuvant. In this context, an adjuvant may be understood as any compound, which is suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response. With other words, when administered, the inventive pharmaceutical composition preferably elicits an innate immune response due to the adjuvant, optionally contained therein. Preferably, such an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal, e.g. an adjuvant protein as defined above or an adjuvant as defined in the following. Particularly preferred as adjuvants suitable for depot and delivery are cationic or polycationic compounds as defined above for the inventive mRNA sequence as vehicle, transfection or complexation agent.
Furthermore, the inventive pharmaceutical composition may comprise one or more additional adjuvants which are suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response, particularly by binding to pathogen-associated molecular patterns (PAMPs). With other words, when administered, the pharmaceutical composition or vaccine preferably elicits an innate immune response due to the adjuvant, optionally contained therein. Preferably, such an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal, e.g. an adjuvant protein as defined above or an adjuvant as defined in the following. According to one embodiment such an adjuvant may be selected from an adjuvant as defined above.
Also such an adjuvant may be selected from any adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal and/or suitable for depot and delivery of the components of the inventive pharmaceutical composition or vaccine. Preferred as adjuvants suitable for depot and delivery are cationic or polycationic compounds as defined above. Likewise, the adjuvant may be selected from the group consisting of, e.g., cationic or polycationic compounds as defined above, from chitosan, TDM, MDP, muramyl dipeptide, pluronics, alum solution, aluminium hydroxide, ADJUMERTM (polyphosphazene); aluminium phosphate gel; glucans from algae; algammulin; aluminium hydroxide gel (alum); highly protein-adsorbing aluminium hydroxide gel; low viscosity aluminium hydroxide gel; AF or SPT (emulsion of squalane (5%), Tween 80 (0.2%), Pluronic L121 (1 .25%), phosphate-buffered saline, pH 7.4); AVRIDINETM (propanediamine); BAY R1005TM ((N-(2-deoxy-2-L-leucylaminob- D- glucopyranosyl)-N-octadecyl-dodecanoyl-amide hydroacetate); CALCITRIOLTM (1 - alpha,25-dihydroxy-vitamin D3); calcium phosphate gel; CAPTM (calcium phosphate nanoparticles); cholera holotoxin, cholera-toxin-A1 -protein-A-D-fragment fusion protein, sub-unit B of the cholera toxin; CRL 1005 (block copolymer P1205); cytokine-containing liposomes; DDA (dimethyldioctadecylammonium bromide); DHEA
(dehydroepiandrosterone); DMPC (dimyristoylphosphatidylcholine); DMPG (dimyristoylphosphatidylglycerol); DOC/alum complex (deoxycholic acid sodium salt); Freund's complete adjuvant; Freund's incomplete adjuvant; gamma inulin; Gerbu adjuvant (mixture of: i) N-acetylglucosaminyl-(P1 -4)-N-acetylmuramyl-L-alanyl-D35 glutamine (GMDP), ii) dimethyldioctadecylammonium chloride (DDA), iii) zinc-L-proline salt complex (ZnPro-8); GM-CSF); GMDP (N-acetylglucosaminyl-(b1 -4)-N-acetylmuramyl-L47 alanyl-D- isoglutamine); imiquimod (1 -(2-methypropyl)-1 H-imidazo[4,5-c]quinoline-4-amine); ImmTher™ (N-acetylglucosaminyl-N-acetylmuramyl-L-Ala-D-isoGlu-L-Ala-glycerol dipalmitate); DRVs (immunoliposomes prepared from dehydration-rehydration vesicles); interferongamma; interleukin-1 beta; interleukin-2; interleukin-7; interleukin-12; ISCOMSTM; ISCOPREP 7.0.3. TM; liposomes; LOXORIBINETM (7-allyl-8-oxoguanosine); LT 5 oral adjuvant {E.coli labile enterotoxin-protoxin); microspheres and microparticles of any composition; MF59TM; (squalenewater emulsion); MONTANIDE ISA 51 TM (purified incomplete Freund's adjuvant); MONTANIDE ISA 720TM (metabolisable oil adjuvant); MPLTM (3-Q-desacyl-4'-monophosphoryl lipid A); MTP-PE and MTP-PE liposomes ((N- acetyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1 ,2-dipalmitoyl-sn-glycero-3- (hydroxyphosphoryloxy))-ethylamide, monosodium salt); MURAMETIDETM (Nac-Mur-L-Ala- D-Gln-OCH3); MURAPALMITINETM and DMURAPALMITINETM (Nac-Mur-L-Thr-D- isoGln-sn-glyceroldipalmitoyl); NAGO (neuraminidase- galactose oxidase); nanospheres or nanoparticles of any composition; NISVs (non-ionic surfactant vesicles); PLEURANTM ( β- glucan); PLGA, PGA and PLA (homo- and co-polymers of lactic acid and glycolic acid; microspheres/nanospheres); PLURONIC L121 TM; PMMA (polymethylmethacrylate); PODDSTM (proteinoid microspheres); polyethylene carbamate derivatives; poly-rA: poly-rU (polyadenylic acid-polyuridylic acid complex); polysorbate 80 (Tween 80); protein cochleates (Avanti Polar Lipids, Inc., Alabaster, AL); STIMULONTM (QS-21 ); Qui l-A (Quil-A saponin); S-28463 (4-amino-otec-dimethyl-2-ethoxymethyl-1 H-imidazo[4,5-c]quinol ine-1 - ethanol); SAF-1 TM ("Syntex adjuvant formulation"); Sendai proteo liposomes and Sendai containing lipid matrices; Span-85 (sorbitan trioleate); Specol (emulsion of Marcol 52, Span 85 and Tween 85); squalene or Robane® (2,6, 10,1 5, 1 9,23-hexamethyltetracosan and 2,6, 1 0, 1 5,1 9,23-hexamethyl-2, 6, 1 0, 14, 1 8,22-tetracosahexane); stearyltyrosine (octadecyltyrosine hydrochloride); Theramid® (N-acetylglucosaminyl-N-acetylmuramyl-L- Ala-D-isoGlu-L-Aladipalmitoxypropylamide); Theronyl-MDP (TermurtideTM or [thr 1 ]-MDP; N-acetylmuramyl-Lthreonyl-D-isoglutamine); Ty particles (Ty-VLPs or virus-like particles); Walter-Reed liposomes (liposomes containing lipid A adsorbed on aluminium hydroxide), and lipopeptides, including Pam3Cys, in particular aluminium salts, such as Adju-phos, Alhydrogel, Rehydragel; emulsions, including CFA, SAF, IFA, MF59, Provax, TiterMax, Montanide, Vaxfectin; copolymers, including Optivax (CRL1 005), LI 21 , Poloaxmer401 0), etc.; liposomes, including Stealth, cochleates, including BIORAL; plant derived adjuvants, including QS21 , Qui l A, Iscomatrix, ISCOM; adjuvants suitable for costimulation including Tomatine, biopolymers, including PLG, PMM, Inulin, microbe derived adjuvants, including Romurtide, DETOX, MPL, CWS, Mannose, CpG nucleic acid sequences, CpG7909, ligands of human TLR 1 -1 0, ligands of murine TLR 1 -1 3, ISS-1 01 8, 35 IC31 , Imidazoquinolines, Ampligen, Ribi529, IMOxine, IRIVs, VLPs, cholera toxin, heat-labile toxin, Pam3Cys, Flagellin, GPI anchor, LNFPIII/Lewis X, antimicrobial peptides, UC-1 V1 50, RSV fusion protein, cdiGMP; and adjuvants suitable as antagonists including CGRP neuropeptide.
Particularly preferred, an adjuvant may be selected from adjuvants, which support induction of a Th1 -immune response or maturation of naive T-cells, such as GM-CSF, IL-1 2, IFNg, any immunostimulatory nucleic acid as defined above, preferably an immunostimulatory RNA, CpG DNA, etc. In a further preferred embodiment it is also possible that the inventive pharmaceutical composition contains besides the antigen-providing mRNA further components which are selected from the group comprising: further antigens or further antigen-providing nucleic acids; a further immunotherapeutic agent; one or more auxiliary substances; or any further compound, which is known to be immunostimulating due to its binding affinity (as l igands) to human Toll-like receptors; and/or an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA).
The inventive pharmaceutical composition can additionally contain one or more auxiliary substances in order to increase its immunogenicity or immunostimulatory capacity, if desired. A synergistic action of the inventive mRNA sequence as defined herein and of an auxiliary substance, which may be optionally contained in the inventive pharmaceutical composition, is preferably achieved thereby. Depending on the various types of auxi liary substances, various mechanisms can come into consideration in this respect. For example, compounds that permit the maturation of dendritic cells (DCs), for example lipopolysaccharides, TNF-alpha or CD40 ligand, form a first class of suitable auxiliary substances. In general, it is possible to use as auxiliary substance any agent that influences the immune system in the manner of a "danger signal" (LPS, GP96, etc.) or cytokines, such as GM-CFS, which allow an immune response to be enhanced and/or influenced in a targeted manner. Particularly preferred auxiliary substances are cytokines, such as monokines, lymphokines, interleukins or chemokines, that further promote the innate immune response, such as IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-1 3, IL-14, IL-1 5, IL-1 6, IL-1 7, IL-1 8, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta or TNF-alpha, growth factors, such as hGH.
Further additives which may be included in the inventive pharmaceutical composition are emulsifiers, such as, for example, Tween®; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet- forming agents; stabilizers; antioxidants; preservatives.
The inventive pharmaceutical composition can also additionally contain any further compound, which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR1 3. In this context it is particularly preferred that the optionally comprised adjuvant component comprises the same inventive m NA as comprised in the inventive pharmaceutical composition as antigen-providing mRNA e.g. mRNA coding for an antigenic peptide or protein of Rabies virus or fragments, variants or derivatives thereof. Despite, the inventive pharmaceutical composition may comprise further components for facilitating administration and uptake of components of the pharmaceutical composition. Such further components may be an appropriate carrier or vehicle, additional adjuvants for supporting any immune response, antibacterial and/or antiviral agents.
Accordingly, in a further embodiment, the inventive pharmaceutical composition furthermore comprises a pharmaceutically acceptable carrier and/or vehicle.
Such a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of a composition comprising the components of the inventive pharmaceutical composition. If the composition is provided in liquid form, the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate etc. buffered solutions. The injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e. the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects. Reference media are e.g. liquids occurring in "in vivd' methods, such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitrd' methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
However, one or more compatible solid or liquid fillers or diluents or encapsulating compounds, which are suitable for administration to a patient to be treated, may be used as well for the pharmaceutical composition according to the invention. The term "compatible" as used here means that these constituents of the inventive pharmaceutical composition are capable of being mixed with the components of the inventive pharmaceutical composition in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the pharmaceutical compostion under typical use conditions. A further component of the inventive pharmaceutical composition may be an immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably immunoglobulins directed against a Rabies virus, eg. Merieux P or Berirab. Preferably, such a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA. Such an immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive antigen-providing mRNA.
Furthermore, in a specific embodiment, additionally to the antigen-providing mRNA further antigens can be included in the inventive pharmaceutical composition and are typically substances such as cells, cell lysates, viruses, attenuated viruses, inactivated viruses, proteins, peptides, nucleic acids or other bio- or macromolecules or fragments thereof. Preferably, antigens may be proteins and peptides or fragments thereof, such as epitopes of those proteins or peptides, preferably having 5 to 15, more preferably 6 to 9, amino acids. Particularly, said proteins, peptides or epitopes may be derived from Glycoprotein G(RAV- G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus or from fragments, variants or derivatives thereof. Further, antigens may also comprise any other biomolecule, e.g., lipids, carbohydrates, etc. Preferably, the antigen is a protein or (poly-) peptide antigen, a nucleic acid, a nucleic acid encoding a protein or (poly-) peptide antigen, a polysaccharide antigen, a polysaccharide conjugate antigen, a lipid antigen, a glycolipid antigen, a carbohydrate antigen, a bacterium, a cell (vaccine), or killed or attenuated viruses. Particularly preferred in this context is the addition of rabies vaccines comprising inactivated virus, as e.g. Rabipur comprising inactived Rabies virus strain Flury-LEP or "inaktivierter (HDC) Tollwutimpfstoff" comprising inactivated Rabies virus strain WISTAR PM/WI 38-1503-3M.
The inventive pharmaceutical composition or vaccine as defined herein may furthermore comprise further additives or additional compounds. Further additives which may be included in the pharmaceutical composition are emulsifiers, such as, for example, Tween®; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet-forming agents; stabilizers; antioxidants; preservatives, RNase inhibitors and/or an anti-bacterial agent or an anti-viral agent. Additionally the inventive pharmaceutical composition may comprise small interfering RNA (siRNA) directed against genes of Rabies virus, e.g. siRNA directed against the gene encoding Glycoprotein G (RAV-G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus.
The inventive pharmaceutical composition typically comprises a "safe and effective amount" of the components of the inventive pharmaceutical composition, particularly of the inventive mRNA sequence(s) as defined herein. As used herein, a "safe and effective amount" means an amount of the inventive mRNA sequence(s) as defined herein as such that is sufficient to significantly induce a positive modification of a disease or disorder or to prevent a disease, preferably rabies as defined herein. At the same time, however, a "safe and effective amount" is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
The inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
According to another particularly preferred aspect, the inventive pharmaceutical composition (or the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein) may be provided or used as a vaccine. Typically, such a vaccine is as defined above for pharmaceutical compositions. Additionally, such a vaccine typically contains the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein. The inventive vaccine may also comprise a pharmaceutically acceptable carrier, adjuvant, and/or vehicle as defined herein for the inventive pharmaceutical composition. In the specific context of the inventive vaccine, the choice of a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered. The inventive vaccine can be administered, for example, systemically or locally. Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes. Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections. More preferably, vaccines may be administered by an intradermal, subcutaneous, or intramuscular route. Inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form. Preferably, the inventive vaccine may be administered by conventional needle injection or needle-free jet injection. In a preferred embodiment the inventive vaccine may be administered by jet injection as defined herein, preferably intramuscularly or intradermal ly, more preferably intradermal ly.
The inventive vaccine can additionally contain one or more auxiliary substances in order to increase its immunogenicity or immunostimulatory capacity, if desired. Particularly preferred are adjuvants as auxiliary substances or additives as defined for the pharmaceutical composition.
In a further aspect, the invention is directed to a kit or kit of parts comprising the components of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine and optionally technical instructions with information on the administration and dosage of the components.
Beside the components of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine the kit may additionally contain a pharmaceutically acceptable vehicle, an adjuvant and at least one further component as defined herein, as wel l as means for administration and technical instructions. The components of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine and e.g. the adjuvant may be provided in lyophilized form. In a preferred embodiment, prior to use of the kit for vaccination, the provided vehicle is than added to the lyophilized components in a predetermined amount as written e.g. in the provided technical instructions. By doing so the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition or vaccine, according to the above described aspects of the invention is provided that can afterwards be used in a method as described above, also. The present invention furthermore provides several applications and uses of the inventive mRNA sequence as defined herei n, of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, of the inventive pharmaceutical composition, of the inventive vaccine, all comprising the inventive mRNA sequence as defined herein or of kits comprising same.
In a further aspect, the invention provides an mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, and a composition, a pharmaceutical composition, a vaccine and a kit, all comprising the mRNA sequence for use in a method of prophylactic (pre-exposure prophylaxis or post-exposure prophylaxis) and/or therapeutic treatment of Rabies virus infections (rabies). Consequently, in a further aspect, the present invention is directed to the first medical use of the inventive mRNA sequence, the inventive composition comprising a plurality of inventive mRNA sequences, the inventive pharmaceutical composition, the inventive vaccine, and the inventive kit as defined herein as a medicament. Particularly, the invention provides the use of an mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof as defined above for the preparation of a medicament. According to another aspect, the present invention is directed to the second medical use of the mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, as defined herein, optionally in form of a composition comprising a plurality of inventive mRNA sequences, a pharmaceutical composition or vaccine, kit or kit of parts, for the treatment of Rabies virus infections (rabies) as defined herein. Particularly, the mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof to be used i n a method as said above is a mRNA sequence formulated together with a pharmaceutical ly acceptable vehicle and an optionally additional adjuvant and an optionally additional further component as defined above e.g. a further antigen or a rabies immune globuline.
In this context the mRNA sequence used for post-exposure treatment of Rabies virus infection according to the invention can be combined with administration of rabies immune globuline.
The inventive mRNA sequence may alternatively be provided such that it is administered for preventing or treating rabies by several doses, each dose containing the inventive mRNA sequence encoding at least one antigenic peptide or protein of Rabies virus, or a fragment, variant or derivative thereof, e.g. the first dose containing at least one mRNA encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G) (or fragments, variants or derivatives thereof) and the second dose containing at least one mRNA sequence encoding at least one antigenic peptide or protein derived from a different antigen of Rabies virus, preferably from the nucleoprotein N (RAV-N) (or fragments, variants or derivatives thereof). By that embodiment, both doses are administered in a staggered way, i.e. subsequently, shortly one after the other, e.g. within less than 10 minutes, preferably less than 2 minutes, and at the same site of the body to achieve the same immunological effect as for administration of one single composition containing both, e.g. the mRNA encoding the glycoprotein G (RAV-G) and the mRNA encoding the nucleoprotein N (RAV-N).
According to a specific embodiment, the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine may be administered to the patient as a single dose or as at least one single dose, respectively. In certain embodiments, the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine may be administered to a patient as a single dose followed by a second dose later and optionally even a third, fourth (or more) dose subsequent thereto etc. In accordance with this embodiment, booster inoculations with the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine may be administered to a patient at specific time intervals, preferably as defined below, following the second (or third, fourth, etc.) inoculation. Preferably, at least one dose of the inventive mRNA sequence, pharmaceutical composition or vaccine is administered, preferably from 1 to 1 0 doses, more preferably from 2 to 7 doses, even more preferably from 2 to 5 doses and most preferably from 3 to 5 doses. In a particularly preferred embodiment, 3 doses are administered. In another embodiment 5 doses are administered. In this context, it is particularly preferred that several doses comprise the same mRNA sequence encoding the same antigenic peptide or protein of Rabies virus, e.g. glycoprotein G (RAV-G). In that embodiment, the doses are given in a specific time period, e.g. 20-30 or 20-60 days. The interval between the administration of two or more doses is preferably from 5 to 120 days, more preferably from 7 to 15 days or 1 5 to 30 days. In a preferred embodiment, the interval between the administration of two or more doses is at least 7days, more preferably 28 days. For example, for post-exposure prophylaxis at least 5 doses of the inventive mRNA sequence or inventive pharmaceutical composition or vaccine can be administered within 20-30 days. As an example, for prophylactic treatment without exposure to the Rabies virus at least 3 doses of the inventive mRNA sequence or the inventive pharmaceutical composition or vaccine can be administered in 20-60 days.
In a preferred embodiment, a single dose of the inventive mRNA sequence, composition or vaccine comprises a specific amount of the mRNA according to the invention. Preferably, the inventive mRNA sequence is provided in an amount of at least 40 pg per dose, preferably in an amount of from 40 to 700 pg per dose, more preferably in an amount of from 80 to 400 pg per dose. More specifically, in the case of intradermal injection, which is preferably carried out by using a conventional needle, the amount of the inventive mRNA sequence comprised in a single dose is typically at least 200 pg, preferably from 200 pg to 1 .000 pg, more preferably from 300 pg to 850 pg, even more preferably from 300 pg to 700 pg. In the case of intradermal injection, which is preferably carried out via jet injection (e.g. using a Tropis device), the amount of the inventive mRNA sequence comprised in a single dose is typically at least 80 pg, preferably from 80 pg to 700 pg, more preferably from 80 pg to 400 pg. Moreover, in the case of intramuscular injection, which is preferably carried out by using a conventional needle or via jet injection, the amount of the inventive mRNA sequence comprised in a single dose is typically at least 80 pg, preferably from 80 pg to 1 .000 pg, more preferably from 80 pg to 850 pg, even more preferably from 80 pg to 700
More specifically, the following specific embodiments are particularly preferred: · the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal I y, in three doses (40 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
• the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (80 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment.
• the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and
56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly by jet injection, in three doses (40 g/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the i nventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermally by jet injection, in three doses (80 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermally by jet injection, in three doses (1 60 g dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intradermal ly by jet injection, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (40 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly in three doses (80 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly, in three doses (640 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (40 g/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on" day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (80 μg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (1 60 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (320 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. the inventive mRNA sequence, or the inventive pharmaceutical composition or vaccine is administered to the patient, preferably intramuscularly by jet injection, in three doses (640 pg/dose), preferably within 20-60 days, e.g. on day 0, 7 and 28 or on day 0, 28 and 56 of the treatment. In certain embodiments, such booster inoculations with the inventive mRNA sequence or inventive pharmaceutical composition or vaccine as disclosed above (second, third etc. vaccination) may utilize an additional compound or component as defined for the inventive mRNA sequence or inventive pharmaceutical composition or vaccine as defined herein.
According to a further aspect, the present invention also provides a method for expression of an encoded antigenic peptide or protein derived from glycoprotein G (RAV-G), nucleoprotein N (RAV-N), phosphoprotein P (RAV-P), matrix protein (RAV-M) or RNA polymerase L (RAV-L) of Rabies virus comprising the steps, e.g. a) providing the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, b) applying or administering the inventive mRNA sequence as defined herein or the inventive composition comprising a plurality of inventive mRNA sequences as defined herein to an expression system, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism. The method may be applied for laboratory, for research, for diagnostic, for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a pharmaceutical composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The method may be carried out in vitro, in vivo or ex vivo. The method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, preferably Rabies as defined herein.
In this context, in vitro is defined herein as transfection or transduction of the inventive mRNA as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein into cells in culture outside of an organism; in vivo is defined herein as transfection or transduction of the inventive mRNA or of the inventive composition comprising a plurality of inventive mRNA sequences into cells by application of the inventive mRNA or of the inventive composition to the whole organism or individual and ex vivo is defined herein as transfection or transduction of the inventive mRNA or of the inventive composition comprising a plurality of inventive mRNA sequences into cells outside of an organism or individual and subsequent application of the transfected cells to the organism or individual.
Likewise, according to another aspect, the present invention also provides the use of the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, preferably for diagnostic or therapeutic purposes, for expression of an encoded antigenic peptide or protein, e.g. by applying or administering the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism. The use may be applied for laboratory, for research, for diagnostic for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive mRNA sequence as defined herein or of the inventive composition comprising a plurality of inventive mRNA sequences as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a pharmaceutical composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The use may be carried out in vitro, in vivo or ex vivo. The use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of Rabies virus infections.
In a further aspect the invention provides a method of treatment or prophylaxis of rabies virus infections comprising the steps:
a) providing the inventive mRNA sequence, the composition comprising a plurality of inventive mRNA sequences, the pharmaceutical composition or the kit or kit of parts comprising the inventive mRNA sequence as defined above;
b) applying or administering the mRNA sequence, the composition, the pharmaceutical composition or the kit or kit of parts to a tissue or an organism;
c) optionally administering rabies immune globuline.
Taken together the invention provides in a certain aspect an mRNA sequence comprising a coding region encoding at least one antigenic peptide or protein of Rabies virus. The inventive mRNA sequence is for use in a method of prophylactic and/or therapeutic treatment of infections caused by Rabies viruses. Accordingly, the invention relates to an mRNA sequence as defined herein for use in a method of prophylactic and/or therapeutic treatment of rabies infections.
In the present invention, if not otherwise indicated, different features of alternatives and embodiments may be combined with each other, where suitable. Furthermore, the term "comprising" shall not be narrowly construed as being limited to "consisting of" only, if not specifically mentioned. Rather, in the context of the present invention, "consisting of" is an embodiment specifically contemplated by the inventors to fall under the scope of "comprising", wherever "comprising" is used herein.
All publications, patents and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.
Figures:
The figures shown in the following are merely illustrative and shall describe the present invention in a further way. These figures shall not be construed to limit the present invention thereto.
Figure 1 : shows the mRNA sequence R2403 according to SEQ ID NO. 24, comprising a
G/C optimized coding region coding for Rabies virus glycoprotein G (RAV-G), the 3'-UTR element muag according to SEQ ID No. 22, a poly (A) sequence consisting of 64 adenosines, a poly(C) sequence consisting of 30 cytosines and a histone stem-loop sequence according to SEQ ID No. 27, as comprised in the RAV-G mRNA vaccine. Figure 2: shows the mRNA sequence R2507 according to SEQ ID NO. 25, comprising a
5'-UTR element comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6, a G/C optimized coding region coding for Rabies virus glycoprotein G (RAV-G), the 3'-UTR element albumin7 according to SEQ ID No. 1 8, a poly (A) sequence consisting of 64 adenosines, a poly(C) sequence consisting of 30 cytosines and a histone stem-loop sequence according to SEQ ID No. 27, as comprised in the RAV-G mRNA vaccine.
Figure 3: shows that transfection of HeLa cells with mRNA R2403 coding for the RAV-G protein leads to the expression of the encoded RAV-G protein on the cell surface and that the protein is recognized by an anti-RAV-G antibody. Construct
R2429 encoding the influenza HA protein of A/Netherlands/602/2009 served as a negative control, as well as untransfected cells. 24 hours post transfection the RAV-G proteins was stained with a rabies specific antibody and FITC-labelled secondary antibody and analysed by FACS as shown in Example 2.
Figure 4: shows that RAV-G mRNA vaccine is immunogenic in mice and induces high titers of neutralizing antibodies comparable to licensed vaccines.
Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine (80 μg of R2403) or Ringer-Lactate (RiLa) as buffer control. Two groups were intramuscularly (i.m.) injected with 1 /1 0 of the human dose of the licensed vaccines Rabipur® and HDC, respectively. All animals received boost injections on day 21 and blood samples were collected on day 35 for the determination of Rabies virus neutralization titers as described in Example 3. (A) The RAV-G mRNA vaccine induced neutralizing antibody titers comparable to the HDC and Rabipur® vaccines, well above the WHO standard of 0.5 l U/ml. The line in the graph represents the median value (n=8 mice/group). (B) The RAV-G mRNA vaccine induced long-lasting virus neutralization titers in mice well above the 0.5 lU/ml.
Figure 5: shows that RAV-G mRNA vaccine induces antigen-specific CD8+ T cel ls.
The experiment was performed as describe in Example 4 and T cel ls were analysed by intracellular cytokine staining for the antigen-specific induction of cytokines. The line in the graph represents the median value (n=8 mice/group).
Figure 6: shows that RAV-G mRNA vaccine induces antigen-specific CD4+ T cells at significantly higher frequencies than Rabipur®.
The experiment was performed as describe in Example 4 and T cells were analysed by intracellular cytokine staining. The line i n the graph represents the median value (n=8 mice/group). Statistical differences between groups were assessed by the Mann Whitney test.
Figure 7: shows that RAV-G mRNA vaccine induces a dose-dependent functional antibody (virus neutralisation antibody) response in C57BL/6 mice, demonstrating dose-response relationship between RAV-G mRNA and the induction of functional antibodies.
The experiment was performed as described in Example 5 and virus neutralization titers (lU/ml) were determined. The line in the graph represents the median value (n=8 mice/group). Statistical analysis: ANOVA (Kruskal- Wallis), **: p 0.01 ; *: p 0.05.
Figure 8: shows that the RAV-G mRNA vaccine protects mice against a lethal rabies virus chal lenge infection.
The experiment was performed as described in Example 6. (A) Survival of rabies infected mice. All mice vaccinated with RAV-G mRNA or Rabipur® were protected against lethal challenge infection without weight loss.
(B) Weight kinetics of rabies infected mice. Several mice vaccinated with the HDC vaccine exhibited weight loss and one mouse reached defined endpoint criteria so that it was terminated before the end of the study.
Figure 9: shows that the RAV-G mRNA vaccine protects mice against a lethal virus challenge infection - Influence of immunization schedule
The experiment was performed as described in Example 6.
(A) Survival of rabies infected mice, (B) Weight kinetics of rabies infected mice. Mice vaccinated three times using RAV-G mRNA in a one or three week intervals between vaccinations were protected against death and weight loss indicating that RAV-G mRNA vaccination is not limited to a fixed immunization schema. In addition, already two vaccinations using RAV-G mRNA were sufficient to protect mice against a lethal challenge infection in terms of protection against death or weight loss.
Figure 10: shows that the RAV-G mRNA vaccine is stable and immunogenic after storage for 6 months at 40°C or 1 month at 60°C.
The experiment was performed as described in Example 7.
(A) Virus neutralization titer. The vaccine was still fully immunogenic after storage for 6 months at temperatures up to 40°C or 1 month at 60°C.
(B) Survival of immunized mice. The vaccine was still fully protective after storage for 6 months up to 40°C or 1 month at 60°C.
Figure 1 1 : shows that the RAV-G mRNA vaccine induces a protective immune response in adult pigs.
The experiment was performed as described in Example 8.
Pigs vaccinated with RAV-G mRNA showed a functional antibody response well above the 0.5 lU/ml WHO standard.
Figure 12: shows that the RAV-G mRNA vaccine induces significant virus neutralization titers in newborn pigs comparable to a benchmark vaccine (Rabipur®).
The experiment was performed as described in Example 9. (A) Kinetics of virus neutralizing titers (mean and standard deviation, SD).
(B) Virus neutralization titers 2 weeks after boost vaccination.
Figure 13: shows that the RAV-G mRNA vaccine induces virus neutralization titers in mice after intramuscular injection. The experiment was performed as described in
Example 1 0.
Figure 14: shows the induction of virus neutralization titers in mice after intradermal vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 1 1 .
Figure 15: shows the induction of virus neutralization titers in domestic pigs after intradermal vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 12.
Figure 16: shows the induction of virus neutralization titers in domestic pigs after intramuscular vaccination with the RAV-G mRNA vaccine. The experiment was performed as described in Example 13.
Examples
The examples shown in the following are merely illustrative and shall describe the present invention in a further way. These examples shall not be construed to limit the present invention thereto.
Example 1 : Preparation of the Rabies mRNA vaccine
1 . Preparation of DNA and mRNA constructs
For the present examples DNA sequences, encoding glycoprotein G (RAV-G) of the Pasteur vaccine strain were prepared and used for subsequent in vitro transcription. The corresponding mRNA sequences RAV-G(GC)-muag-A64-C30-histoneSL (R2403) and 32L-RAV-G(GC)-albumin7-A64-C30-histoneSL (R2507) are shown in Figure 1 and 2 according to SEQ. ID No. 24 and 25.
2. In vitro transcription
The respective DNA plasmids prepared according to paragraph 1 were transcribed in vitro using T7 polymerase in the presence of a CAP analogue (m7GpppG). Subsequently the mRNA was purified using PureMessenger® (CureVac, Tiibingen, Germany; WO 2008/077592A1 ).
The mRNA sequence RAV-G(GC)-muag-A64-C30-histoneSL (R2403; SEQ ID NO:24) comprises in 5'- to 3'-direction:
a. ) a 5'-CAP structure consisting of m7GpppN; b. ) a G/C maximized coding region encoding the full-length protein of RAV-G of the
Pasteur vaccine strain according to SEQ ID No. 1 ; c. ) a 3'-UTR element derived from a alpha globin gene, comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22; d. ) a poly(A) sequence, comprising 64 adenosines; e. ) a poly(C) sequence, comprising 30 cytosines; and f.) a histone-stem-loop structure, comprising the RNA sequence according to SEQ ID No 27.
The term "R2403", as used herein, refers to the mRNA sequence, which is defined by the sequence according to SEQ ID NO:24. The "R2403" mRNA may be provided in lyophilised form, which is preferably used for storage and/or transport of the inventive mRNA sequence or may be provided in the solved form in the appropriate liquid. Before administration to a subject, the mRNA according to SEQ ID NO:24, if provided in lyophilised form, is typically reconstituted in an appropriate liquid as defined herein, preferably in Ringer-Lactate, in order to obtain a liquid formulation.
The mRNA sequence 32L-RAV-G(GC)-albumin7-A64-C30-histoneSL (R2507) comprises in 5'- to 3'-direction:
a. ) a 5'-CAP structure, consisting of m7GpppN; b. ) a 5'-UTR element comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6; c. ) a G/C-maximized coding region encoding the full-length protein of RAV-G of the
Pasteur vaccine strain according to SEQ ID No. 1 ; d. ) a 3'UTR element comprising the corresponding RNA sequence of a nucleic acid sequence according to SEQ ID NO. 18; e. ) a poly(A) sequence, comprising 64 adenosines; f. ) a poly(C) sequence, comprising 30 cytosines; and g. ) a histone-stem-loop structure, comprising the RNA sequence according to SEQ
ID No 27.
Reagents
Complexation Reagent: protamine
Preparation of the vaccine
The mRNA R2403 or R2507 were complexed with protamine by addition of protamine to the mRNA in the ratio (1 :2) (w/w) (adjuvant component). After incubation for 10 min, the same amount of free mRNA R2403 or R2507 used as antigen-providing mRNA was added.
Example 2: In vitro characterization of mRNA encoding Rabies virus G protein (RAV-G)
HeLa cells were seeded in a 6-well plate at a density of 300 000 cells/well in cell culture medium (RPMI, 10% FCS, 1 % L-Glutamine, 1 % Pen/Strep) 24h prior to transfection. HeLa cells were transfected with 5 μg of RAV-G encoding mRNA (R2403) or influenza HA protein of A/Netherlands/602/2009 encoding mRNA (R2429) as negative control using Lipofectamine 2000 (Invitrogen) and stained 24 hours post transfection with a rabies virus specific antibody (HyTest Ltd; #1 1/06-R7-C5) and FITC labelled goat anti-mouse IgG antibody (Invitrogen, #871942A) and analysed by flow cytometry (FACS). The flow cytometry data are evaluated quantitatively by Flowjo software. Figure 3 demonstrates that the RAV-G protein is, as expected for the Rabies G protein, expressed on the surface of transfected cells and can be recognized by an anti-rabies antibody.
Example 3: Induction of a humoral immune response by the RAV-G mRNA vaccine Immunization
On day zero, BALB/c mice were intradermally (i.d.) injected with the mRNA vaccine comprising mRNA coding for Rabies virus glycoprotein G (RAV-G) (R2403 according to Example 1 ; 80 μg/mouse/vacci nation day) or Ringer-lactate (RiLa) as buffer control. Two control groups were intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccines Rabipur® (Novartis) and HDC (human diploid cell vaccine, Sanofi Pasteur MSD GmbH), respectively. All animals received boost injections on day 21 and blood samples were collected on day 35 for the determination of virus neutralization titers.
To establish a long term kinetic of the anti-RAV-G immune response, blood samples were taken from group 1 after 1 5, 29, 38 and 48 weeks and virus neutralization titers were determined. Table 1 : Animal groups
The licensed rabies vaccines Rabipur® (Novartis) and HDC (Human diploid cells, Sanofi Pasteur MSD GmbH) comprise inactivated Rabies virus.
Virus neutralization test
Detection of the virus neutral izing antibody response (specific B-cel l immune response) was carried out by a virus neutralisation assay. The result of that assay is referred to as virus neutralization titer (VNT). According to WHO standards, an antibody titer is considered protective if the respective VNT is at least 0.5 l U/ml. Therefore, blood samples were taken from vaccinated mice on day 35 and from vaccinated humans on day 42 or as i ndicated after vaccination and sera were prepared. These sera were used in fluorescent antibody virus neutralisation (FAVN) test using the cell culture adapted challenge virus strain (CVS) of rabies virus as recommended by the OIE (World Organisation for Animal Health) and first described in Cliquet F., Aubert M. & Sagne L. (1 998); J. Immunol. Methods, 21 2, 79-87. Shortly, heat inactivated sera will be tested as quadruplicates in serial two-fold dilutions as quadruplicates for their potential to neutralise 1 00 TCI D5o (tissue culture infectious doses 50 % ) of CVS in 50 μΙ of volume. Therefore sera dilutions are incubated with virus for 1 hour at 37°C (in humid incubator with 5% CO2) and subsequently trypsi nized BHK-21 cel ls are added (4x105 cells/ml; 50 μΙ per wel l). Infected cell cultures are incubated for 48 hours in humid incubator at 37°C and 5 % CO2. Infection of cells is analysed after fixation of cel ls using 80% acetone at room temperature using FITC anti-rabies conjugate. Plates were washed twice using PBS and excess of PBS was removed. Cell cultures are scored positive or negative for the presence of rabies virus. Negative scored cells in sera treated wells represent neutralization of rabies virus. Each FAVN tests includes WHO or OIE standard serum (positive reference serum) that serves as reference for standardisation of the assay. Neutralization activity of test sera is calculated with reference to the standard serum provided by the WHO and displayed as International Units/ml (lU/ml).
Results
As can be seen in Figure 4A, the RAV-G mRNA vaccine (R2403) induces neutralizing antibody titers comparable to the HDC and Rabipur® vaccines, well above the WHO standard of 0.5 lU/ml.
As can be seen from Figure 4B, the RAV-G mRNA vaccine induces long-lasting rabies virus neutralization titers in mice.
Example 4: Induction of a cellular immune response by the RAV-G mRNA vaccine Immunization
On day zero, BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 (80 μg/mouse/vaccination/day) or Ringer-lactate (RiLa) as buffer control. A control group was intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccine Rabipur®. All animals received boost injections on day 21 . Serum and spleens were collected (n=8 on day 28, n=8 on day 35) for the analysis of antigen-specific T cells.
Table 2: Animal groups
Intracellular cytokine staining Splenocytes from vaccinated and control mice were isolated according to a standard protocol. Briefly, isolated spleens were grinded through a cell strainer and washed in PBS/1 %FBS followed by red blood cel l lysis. After an extensive washing step with PBS/1 %FBS splenocytes were seeded into 96-well plates (2x1 06 cells/well) and kept overnight at 4°C. The next day cells were stimulated with the RAV-G peptide library (J PT) that comprised the amino acid sequence of the Rabies G protein from Pasteur vaccine strain of Rabies virus according to SEQ ID No. 1 displayed as 1 5 amino acid peptides with an overlap of 1 1 amino acids between adjacent peptides and 2.5 μg/ml of an anti-CD28 antibody (BD Biosciences) for 6 hours at 37°C in the presence of the mixture of GolgiPlug™/GolgiStop™ (Protein transport inhibitors containing Brefeldin A and Monensin, respectively; BD Biosciences). After stimulation cells were washed and stained for intracellular cytokines using the Cytofix/Cytoperm reagent (BD Biosciences) according to the manufacturer's instructions. The fol lowing antibodies were used for staining: CD8-PECy7 (1 :200), CD3-FITC (1 :200), IL2-PerCP-Cy5.5 (1 :100), TNFa-PE (1 : 100), IFNv-APC (1 :1 00) (eBioscience), CD4-BD Horizon V450 (1 :200) (BD Biosciences) and incubated with FcyR- block diluted 1 :1 00. Aqua Dye was used to distinguish live/dead cells (Invitrogen). Cells were collected using a Canto II flow cytometer (Beckton Dickinson). Flow cytometry data were analysed using Flowjo software (Tree Star, Inc.). Statistical analysis was performed using GraphPad Prism software, Version 5.01 . Statistical differences between groups were assessed by the Mann Whitney test.
Results As can be seen from Figure 5, the RAV-G mRNA vaccine (R2403) induced IFNy positive, TNFa positive and IFNy/TNFa double-positive multifunctional CD8+ T cells at comparable frequencies as the Rabipur® vaccine.
As can be seen from Figure 6, the RAV-G mRNA vaccine (R2403) induced IFNy positive, TNFa positive and IFNy TNFa double-positive multifunctional CD4+ T cells at significantly higher frequencies than the Rabipur® vaccine which comprises the whole inactivated Rabies virus.
Example 5: Induction of a dose-dependent humoral immune response by the RAV-G mRNA vaccine in C57BL/6 mice
Immunization
On day zero, C57BL/6 mice were intradermally (i.d.) injected with different doses of the RAV-G mRNA vaccine R2403 or Ringer-lactate (RiLa) as buffer control as shown in Table 3. Two groups were intramuscularly (i.m.) injected with 1 /1 0 of the human dose of the licensed vaccines Rabipur® and HDC, respectively. All animals received boost injections on day 21 . Blood samples were taken on day 35 and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3.
Table 3: Animal groups
Group Strain Number Route Vaccine Vaccination
sex of mice volume dose schedule
1 C57BL/6 8 i.d. R2403 dO: prime, d21 :
Female 2x 50 μΙ 80 μδ boost
d35:blood collection
2 C57BL/6 8 i.d. R2403 dO: prime, d21 :
Female 2x 50 μΙ 40 μ% boost
d35:blood collection
3 C57BL/6 8 i.d. R2403 dO: prime, d21 :
Female 2x 50 μΙ 0 μ% boost
d35:blood collection
4 C57BL/6 8 i.d. R2403 dO: prime, d21 :
Female 2x 50 μΙ 1 0 μδ boost
d35:blood collection
5 C57BL/6 8 i.d. R2403 dO: prime, d21 :
Female 2x 50 μΙ boost
d35:blood collection
6 C57BL/6 8 i.m. HDC inactivated dO: prime, d21 :
Female 4x 25 μΙ (0.1 of human dose) boost
d35:blood collection
7 C57BL/6 8 i.m. Rabipur® dO: prime, d21 :
Female 4x 25 μΙ (0.1 of human dose) boost
d35:blood collection
8 C57BL/6 8 i.d. 100% Ringer dO: prime, d21 :
Female 2x 50 μΙ Lactate (RiLa) buffer boost
d35:blood collection Results
As can be seen from Figure 6, the RAV-G mRNA vaccine induces a dose-dependent antibody response. Doses of 20 μg, 40 μg and 80 μg induced significantly higher virus neutralizing antibody titers compared to 0.1 human dose of the HDC vaccine.
Example 6: Rabies virus challenge infection of mice Immunization
Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 according to the schedule shown in Table 3 or Ringer-lactate (RiLa) as buffer control. Two control groups were intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccines Rabipur® and HDC, respectively. Sixteen days after the last immunization the animals were infected using a 40-fold LD50 dose of the CVS strain of Rabies virus intracranial ly (i.e.). Mice were monitored for specific symptoms of Rabies disease and body weight development.
Table 4: Animal groups
Results
As can be seen from Figure 8A, the RAV-G mRNA vaccine protected all mice against a lethal rabies virus challenge infection. All mice vaccinated with RAV-G mRNA or Rabipur® were protected against a lethal challenge infection without weight loss.
As can be seen from Figure 8B, several mice vaccinated with the HDC vaccine exhibited weight loss and one diseased mouse in the HDC group reached defined endpoint criteria and was therefore terminated before the end of the study.
As can be seen from Figure 9A, two vaccinations with the RAV-G mRNA vaccine (Prime / Boost in three week interval) were sufficient to fully protect mice against a lethal challenge infection with rabies virus. Example 7: Induction of a humoral immune response by the RAV-G mRNA vaccine after storage
To test the stability of the RAV-G mRNA vaccine, samples were stored at 5°C, 25° and 40°C for 6 months and at 60°C for one month. Subsequently, mice were vaccinated with these samples and their immunogenic and protective potential was evaluated.
Immunization
Female BALB/c mice were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 according to the schedule shown in Table 5 or Ringer-lactate (RiLa) as buffer control. A control group was intramuscularly (i.m.) injected with 1/10 of the human dose of the licensed vaccine HDC (stored as recommended by the manufacture at 2-8°C). Two weeks after the last vaccination blood samples were collected and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3. Sechs weeks after the last immunization the animals were infected using 25-fold LD50 of CVS strain of Rabies virus intracranially (i.e.). Mice were monitored for specific symptoms of Rabies disease and body weight development.
Table 5: Animal groups
Results
As can be seen from Figure 10A, the RAV-G mRNA vaccine is stable and immunogenic after storage for 6 months up to 40°C or 1 month at 60°C. In addition, the vaccine is still fully protective as demonstrated in a lethal challenge infection (10B)
Example 8: Induction of a humoral immune response by the RAV-G mRNA vaccine in pigs Immunization
Two groups of pigs (Hungarian large white pig, 6 to 8 weeks old, female; n=5) were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2507 or Ringer-lactate (RiLa) as buffer control according to the schedule shown in Table 6. One week before (preimmune serum) and 2, 3 and 5 weeks after the first vaccination blood samples were collected and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3. Table 6: Animal groups
Results
As can be seen from Figure 1 1 , the RAV-G mRNA vaccine induces an immune response after prime / boost vaccination well above the 0.5 lU/ml WHO standard.
Example 9: Induction of virus neutralization titers by the RAV-G mRNA vaccine in newborn pigs is comparable to a benchmark vaccine (Rabipur®)
Immunization
On day zero, 3 to 4 day old piglets (German domestic pig, of both genders) from two litters were intradermally (i.d.) injected with the RAV-G mRNA vaccine R2403 and an unrelated control mRNA vaccine (R2402 encoding the HA protein of influenza H5N1 ) as shown in Table 7. A third group was intramuscularly (i.m.) injected with one human dose of the licensed vaccine Rabipur®. All animals received boost injections on day 21 . Blood samples were taken on day 0 (preimmune serum) and days 21 , 28, 35, 49 and 70. Sera were prepared and analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3. Table 7: Animal groups
Results
As can be seen from Figure 12, the RAV-G mRNA vaccine induces virus neutralizing antibodies after prime-boost vaccination of new born pigs, well above the 0.5 lU/ml WHO standard.
Example 10: Induction of virus neutralization titers by the RAV-G mRNA vaccine after intramuscular immunization in mice
Immunization
Female BALB/c mice were intramuscularly (i.m.; M. tibialis) injected with the RAV-G mRNA vaccine R2507 or Ringer- lactate (RiLa) as buffer control according to the schedule shown in Table 8. One week after the last vaccination blood samples were collected and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3. Table 8: Animal groups
Results
As can be seen from Figure 1 3, the RAV-G mRNA vaccine induces virus neutralization titers in mice after intramuscular injection well above the 0.5 lU/ml WHO standard.
Example 1 1 : Induction of a humoral immune response by the RAV-G mRNA vaccine in mice
Immunization
On day zero, BALB/c mice were intradermally (i.d.) injected with the mRNA vaccine comprising mRNA coding for Rabies virus glycoprotein G (RAV-G) (according to Example 1 ; 80 ^g/mouse/vacci nation day). All animals received boost injections on day 7 and 21 . To establish a long term kinetic of the anti-RAV-G immune response blood samples were collected on day 0, 21 and 35 for the determination of virus neutralization titers.
Virus neutralization test
The virus neutralization test was performed as described in Example 3. Results
As can be seen in Figure 14, the RAV-G mRNA vaccine (R2403) induces already after two vaccinations neutralizing antibody titers, well above the WHO standard of 0.5 lU/ml. Example 12: Induction of a humoral immune response by the RAV-G mRNA vaccine in domestic pigs
Immunization
Two groups of pigs (Hungarian large white pig, 6 to 8 weeks old, female; n=5) were intradermally (i.d.) injected on day 0 and 14 with the RAV-G mRNA vaccine R2507 or Ringer-lactate (RiLa) as buffer control. One week before (preimmune serum) the first vaccination (day 0) and on day 14 and 21 after the first vaccination blood samples were collected and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3.
Results
As can be seen from Figure 1 5, only one vaccination with the RAV-G mRNA vaccine is sufficient to reach the WHO standard of 0.5 lU/ml neutralizing antibody titers in domestic pigs.
Example 13: Induction, of a humoral immune response by the RAV-G mRNA vaccine in domestic pigs Immunization
Domestic pigs (Hungarian large white pig, 6 to 8 weeks old, female; n=5) were intramuscularly (i.m.) injected on day 1 , 8 and 29 with the RAV-G mRNA vaccine R2403. Blood samples were collected on day 1 , 8, 15, 29, 36, 43, 57 and 1 1 5 and sera were analysed in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3.
Results
As can be seen from Figure 1 6, intramuscular vaccination with the RAV-G mRNA vaccine is able to induce neutralizing antibody titers above the WHO standard of 0.5 lU/ml in domestic pigs. The long-term kinetic shows that even 1 15 days after the last vaccination the neutralizing antibody titers are above the WHO standard of 0.5 lU/ml. Example 14: Induction of a humoral immune response by the RAV-G mRNA vaccine in humans
Immunization
Preliminary results obtained in an ongoing clinical trial (phase I) demonstrate safety as well as efficacy of the vaccine according to the invention. In the clinical study, human volunteers were intradermally injected via jet injection using a Tropis device on day 0, 7 and 28 with the RAV-G mRNA vaccine R2403. The mRNA was prepared as described in Example 1 herein, i.e. mRNA complexed with protamine in a ratio of 2:1 (w/w) was mixed with an equal amount of free mRNA. On each of the three vaccination days, 80 pg of mRNA were administered.
In order to assess the safety profile of the vaccine according to the invention, subjects were monitored after administration (vital signs, vaccination site tolerability assessments, hematologic analysis after the second and third injection). The preliminary results obtained in the ongoing clinical study suggest that immunization with the mRNA according to the invention is we 11 -tolerated in humans.
The efficacy of the immunization was analysed by determination of virus neutralizing titers (VNT) in sera from six subjects. To this end, blood samples were collected on day 0 as baseline and on day 42. Sera were analysed for virus neutralizing antibodies in the fluorescent antibody virus neutralisation (FAVN) test as described in Example 3. The results are summarized in Table 9. Table 9: Virus neutralizing titers after immunization of human subjects
In five out of six subjects (subject no. 1 , 2, 4, 5 and 6), a virus neutralizing titer of at least 0.5 lU/ml was detected on day 42. According to the WHO standard, a protective antibody response has thus been achieved in these subjects, demonstrating the efficacy of the immunization with the mRNA according to the invention.
Conclusion:
According to preliminary results from the ongoing clinical trial, the use of the mRNA according to the invention for immunization of human subjects has a favourable safety profile. The efficacy of the approach has been demonstrated by these preliminary studies with a protective antibody response (VNT > 0.5 lU/ml) achieved in five out of six investigated subjects on day 42.

Claims

Claims mRNA sequence comprising a coding region, encoding at least one antigenic peptide or protein derived from the glycoprotein G (RAV-G), nucleoprotein N (RAV- N), phospoprotein P (RAV-P), matrix protein M (RAV-M) or RNA polymerase L (RAV- L) of Rabies virus or a fragment, variant or derivative thereof;
wherein the G/C content of the coding region is increased compared with the G/C content of the coding region of the wild type mRNA, and wherein the coded amino acid sequence of said GC-enriched mRNA is preferably not being modified compared with the coded amino acid sequence of the wild type mRNA.
The mRNA sequence according to claim 1 , wherein the coding region encodes the full-length protein of glycoprotein G (RAV-G) of Rabies virus.
The mRNA sequence according to any of claims 1 to 2, wherein the antigenic peptide or protein is derived from a Rabies vaccine strain, particularly of the Pasteur vaccine strain or of the Flury-LEP vaccine strain.
The mRNA sequence according to any of claims 1 to 3 comprising additionally a) a 5 '-CAP structure,
b) a poly(A) sequence,
c) and optionally a poly (C) sequence.
The mRNA sequence according to claim 4, wherein the poly(A) sequence comprises a sequence of about 25 to about 400 adenosine nucleotides, preferably a sequence of about 50 to about 400 adenosine nucleotides, more preferably a sequence of about 50 to about 300 adenosine nucleotides, even more preferably a sequence of about 50 to about 250 adenosine nucleotides, most preferably a sequence of about 60 to about 250 adenosine nucleotides.
The mRNA sequence according to any of claims 1 to 5 comprising additionally at least one histone stem-loop.
The mRNA sequence according to claim 6, wherein the at least one histone stem- loop is selected from following formulae (I) or (II): formula (I) (stem-loop sequence without stem bordering elements):
steml loop stem2 ula (II) (stem-loop sequence with stem bordering elements):
N1-6 [N0-2GN3-5] [No-4(U/T)No-4] [N3-5CN0-2]
-J J ^ steml steml stem2 stem2 bordering element bordering element wherein: steml or stem2 bordering elements N 1 -6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof; steml [N0-2GN3-5] is reverse complementary or partially reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof, and wherein G is guanosine or an analogue thereof, and may be optionally replaced by a cytidine or an analogue thereof, provided that its complementary nucleotide cytidine in stem2 is replaced by guanosine;
4(U/T)No-4] is located between elements steml and stem2, and is a consecutive sequence of 3 to 5 nucleotides, more preferably of 4 nucleotides; wherein each N0-4 is independent from another a consecutive sequence of 0 to 4, preferably of 1 to 3, more preferably of 1 to 2 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; and wherein U/T represents uridine, or optionally thymidine;
stem2 [N3-5CN0-2] is reverse complementary or partially reverse complementary with element steml , and is a consecutive sequence between of 5 to 7 nucleotides; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleotide guanosine in stem! is replaced by cytidine;
wherein
steml and stem2 are capable of base pairing with each other
forming a reverse complementary sequence, wherein base pairing may occur between steml and stem2, or
forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between steml and stem2.
The mRNA sequence according to claim 7, wherein the at least one histone stem- loop is selected from at least one of following formulae (la) or (Ma):
[N0-1 GN3-5] [Ni-3(U/T)IMo-2] [N3-5CN0-1 ]
' γ J > ¥ <
steml loop stem2
formula (la) (stem-loop sequence without stem bordering elements)
N2-5 [N0-1 GN3-5] [Ni.3(U T)No-2] [N3-5CN0-1 ] N2-5
. v , · , · ^ steml steml loop stem2 stem2
bordering element bordering element
formula (Ha) (stem-loop sequence with stem bordering elements)
9. The mRNA sequence according to any of claims 1 to 8 comprising additionally a 3'- UTR element. The mRNA sequence according to claim 9, wherein the at least one 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of a gene providing a stable mRNA or from a homolog, a fragment or a variant thereof.
The mRNA sequence according to claim 1 0, wherein the 3'UTR element comprises or consists of a nucleic acid sequence derived from a 3'UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a β-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, or from a homolog, a fragment or a variant thereof.
The mRNA sequence according to claim 1 1 , wherein the 3'-UTR element comprises or consists of a nucleic acid sequence derived from a 3'UTR of a-globin gene, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22, a homolog, a fragment, or a variant thereof;
The mRNA sequence according to any of claims 9 to 12; wherein the mRNA sequence comprises, preferably in 5'- to 3'-direction:
a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; c. ) a 3'-UTR element comprising or consisting of a nucleic acid sequenc which is derived from a alpha globin gene, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 22, a homolog, a fragment or a variant thereof; d. ) a poly(A) sequence, preferably comprising 64 adenosines; e. ) a poly(C) sequence, preferably comprising 30 cytosines; and f. ) a histone-stem-loop, preferably comprising the corresponding RNA sequence to the nucleic acid sequence according to SEQ ID No 23.
14. The mRNA sequence according to claim 13, wherein the mRNA sequence comprises the RNA sequence according to SEQ ID No. 24. The mRNA sequence according to claim 9, wherein the at least one 3'UTR element comprises or consist of a nucleic acid sequence which is derived from the 3'UTR of a vertebrate albumin gene or from a variant thereof, preferably from the 3'UTR of a mammalian albumin gene or from a variant thereof, more preferably from the 3'UTR of a human albumin gene or from a variant thereof, even more preferably from the 3'UTR of the human albumin gene according to GenBank Accession number NM_000477.5, or from a fragment or variant thereof.
The mRNA sequence according to claim 1 5, wherein the 3'UTR element is derived from a nucleic acid sequence according to SEQ ID NO. 1 8, preferably from a corresponding RNA sequence, a homolog, a fragment or a variant thereof.
The mRNA sequence according to any of claims 1 to 1 6 comprising additionally a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'UTR of a TOP gene preferably from a corresponding RNA sequence, a homolog, a fragment, or a variant thereof, preferably lacking the 5'TOP motif.
The mRNA sequence according to claim 1 7, wherein the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal protein, preferably from a corresponding RNA sequence or from a homolog, a fragment or a variant thereof, preferably lacking the 5'TOP motif.
The mRNA sequence according to claim 1 8, wherein the 5'UTR element comprises or consists of a nucleic acid sequence which is derived from a 5'UTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog, a fragment or variant thereof, preferably lacking the 5'TOP motif and more preferably comprising or consisting of a corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6.
20. The mRNA sequence according to claim 1 9; wherein the mRNA sequence comprises, preferably in 5'- to 3'-direction: a.) a 5'-CAP structure, preferably m7GpppN; b. ) a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, preferably comprising or consisting of the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO. 1 6 , a homolog, a fragment or a variant thereof; c. ) a coding region encoding at least one antigenic peptide or protein of Rabies virus, preferably derived from the glycoprotein G (RAV-G) of Rabies virus; d. ) a 3'UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable mRNA, preferably comprising or consisting of the corresponding RNA sequence of a nucleic acid sequence according to SEQ ID NO. 1 8, a homolog, a fragment or a variant thereof; e. ) a poly(A) sequence preferably comprising 64 adenosines; f. ) a poly(C) sequence, preferably comprising 30 cytosines; and g. ) a histone-stem-loop, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID No 23.
The mRNA sequence according to claim 20, wherein the mRNA sequence comprises the RNA sequence according to SEQ ID No. 25.
The mRNA sequence according to claims 1 to 21 , wherein the mRNA sequence is associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w:w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w) of mRNA to cationic or polycationic compound and/or with a polymeric carrier; or optionally in a nitrogen/phosphate ratio of mRNA to cationic or polycationic compound and/or polymeric carrier in the range of about 0.1 -1 0, preferably in a range of about 0.3-4 or 0.3-1 , and most preferably in a range of about 0.5-1 or 0.7-1 , and even most preferably in a range of about 0.3-0.9 or 0.5-0.9.
The mRNA sequence according to claim 22, wherein the mRNA sequence is associated or complexed with a cationic protein or peptide, preferably protamine.
24. A composition comprising a plurality or more than one of mRNA sequences each according to any of claims 1 to 23. 25. Pharmaceutical composition comprising an mRNA sequence as defi ned according to any of claims 1 to 23 or a composition as defined according to claim 24 and optionally a pharmaceutically acceptable carrier.
26. Pharmaceutical composition according to claim 25, wherein the mRNA sequence is complexed at least partially with a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides and most preferably protamine.
27. Pharmaceutical composition according to claim 26, wherein the ratio of complexed mRNA to free mRNA is selected from a range, of about 5: 1 (w/w) to about 1 : 1 0
(w/w), more preferably from a range of about 4: 1 (w/w) to about 1 :8 (w/w), even more preferably from a range of about 3 :1 (w/w) to about 1 :5 (w/w) or 1 :3 (w/w), and most preferably the ratio of complexed mRNA to free mRNA is selected from a ratio of 1 : 1 (w/w).
28. Kit or kit of parts comprising the components of the mRNA sequence as defined according to any of claims 1 to 23, the composition as defined according to claim 24, the pharmaceutical composition as defined according to any of claims 25 to 27 and optionally technical instructions with information on the administration and dosage of the components.
29. mRNA sequence as defined according to any of claims 1 to 23, composition as defined according to claim 24, pharmaceutical composition as defined according to any of claims 25 to 27, and kit or kit of parts as defined according to claim 28 for use as a medicament.
30. mRNA sequence as defined according to any of claims 1 to 23, composition as defined according to claim 24, pharmaceutical composition as defined according to any of claims 25 to 27, and kit or kit of parts as defined according to claim 28 for use in the treatment or prophylaxis of Rabies virus infections. 31 mRNA sequence, composition, pharmaceutical composition and kit or kit of parts for use according to claim 30, wherein the treatment is a post-exposure prophylaxis. mRNA sequence, composition, pharmaceutical composition and kit or kit of parts for use according to claim 31 , wherein the treatment is combined with administration of rabies immue globuline. mRNA sequence, composition, pharmaceutical composition and kit or kit of parts for use according to any of claims 29 to 32, wherein the mRNA sequence, the composition, the pharmaceutical composition or the kit or kit of parts is administered by subcutaneous, intramuscular or intradermal injection, preferably by intramuscular or intradermal injection, more preferably by intradermal injection. mRNA sequence, composition, pharmaceutical composition and kit or kit of parts for use according to claim 33, wherein the injection is carried out by using conventional needle injection or jet injection, preferably by using jet injection.
A method of treatment or prophlaxis of rabies virus infections comprising the steps: a) providing the mRNA sequence as defined according to any of claims 1 to 23, the composition as defined according to claim 24, the pharmaceutical composition as defined according to any of claims 25 to 27, or the kit or kit of parts as defined according to claim 28;
b) applying or administering the mRNA sequence, the composition, the
pharmaceutical composition or the kit or kit of parts to a tissue or an organism;
c) optional ly administering rabies immune globuline.
The method according to claim 35, wherein the mRNA sequence, the composition, the pharmaceutical composition or the kit or kit of parts is administered to the tissue or to the organism by subcutaneous, intramuscular or intradermal injection, preferably by intramuscular or intradermal injection, more preferably by intradermal injection.
37. The method according to claim 36, wherein the injection is carried out by using conventional needle injection or jet injection, preferably by using jet injection.
EP14755333.3A 2013-08-21 2014-08-21 Rabies vaccine Withdrawn EP3035961A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP14755333.3A EP3035961A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
EP18191308.8A EP3461498A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
EP22151797.2A EP4043032A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP2013002517 2013-08-21
EP14755333.3A EP3035961A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
PCT/EP2014/002298 WO2015024665A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP18191308.8A Division EP3461498A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
EP22151797.2A Division EP4043032A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Publications (1)

Publication Number Publication Date
EP3035961A1 true EP3035961A1 (en) 2016-06-29

Family

ID=56024445

Family Applications (3)

Application Number Title Priority Date Filing Date
EP18191308.8A Withdrawn EP3461498A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
EP14755333.3A Withdrawn EP3035961A1 (en) 2013-08-21 2014-08-21 Rabies vaccine
EP22151797.2A Pending EP4043032A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP18191308.8A Withdrawn EP3461498A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP22151797.2A Pending EP4043032A1 (en) 2013-08-21 2014-08-21 Rabies vaccine

Country Status (1)

Country Link
EP (3) EP3461498A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4550115B2 (en) * 2004-08-03 2010-09-22 ジーンアート・アクチエンゲゼルシャフト Method for regulating gene expression by changing CpG content
DE102006061015A1 (en) 2006-12-22 2008-06-26 Curevac Gmbh Process for the purification of RNA on a preparative scale by HPLC
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
ES2558106T3 (en) 2010-07-30 2016-02-02 Curevac Ag Formation of nucleic acid complexes with disulfide-cross-linked cationic components for transfection and immunostimulation
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
WO2012116715A1 (en) * 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
WO2012116714A1 (en) * 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
SG11201405545XA (en) 2012-03-27 2014-11-27 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
AMANN R ET AL: "A new rabies vaccine based on a recombinant ORF virus (parapoxvirus) expressing the rabies virus glycoprotein", JOURNAL OF VIROLOGY (ONLINE), AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 87, no. 3, 1 February 2013 (2013-02-01), pages 1618 - 1630, XP002753921, ISSN: 1098-5514, [retrieved on 20121121], DOI: 10.1128/JVI.02470-12 *
HILDEGUND C. J. ERTL ET AL: "Novel Vaccines to Human Rabies", PLOS NEGLECTED TROPICAL DISEASES, vol. 3, no. 9, 29 September 2009 (2009-09-29), US, pages e515, XP055450808, ISSN: 1935-2727, DOI: 10.1371/journal.pntd.0000515 *
See also references of WO2015024665A1 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents

Also Published As

Publication number Publication date
EP3461498A1 (en) 2019-04-03
EP4043032A1 (en) 2022-08-17

Similar Documents

Publication Publication Date Title
US11369694B2 (en) Rabies vaccine
US11739125B2 (en) Respiratory syncytial virus (RSV) vaccine
US20210401966A1 (en) Nucleic acid molecules and uses thereof
US20170326225A1 (en) Ebolavirus and marburgvirus vaccines
US20210162037A1 (en) Influenza mrna vaccines
EP3035960B1 (en) Respiratory syncytial virus (rsv) vaccine
EP4043032A1 (en) Rabies vaccine
US11965000B2 (en) Respiratory syncytial virus (RSV) vaccine

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20151230

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
RIN1 Information on inventor provided before grant (corrected)

Inventor name: KRAMPS, THOMAS

Inventor name: PETSCH, BENJAMIN

Inventor name: STITZ, LOTHAR

Inventor name: SCHNEE, MARGIT

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170718

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20180911